Language selection

Search

Patent 2070979 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent: (11) CA 2070979
(54) English Title: SEQUENCE SPECIFIC DNA BINDING BY P53
(54) French Title: LIAISON D'ADN DE SEQUENCE SPECIFIQUE PAR P53
Status: Expired
Bibliographic Data
(51) International Patent Classification (IPC):
  • C12Q 1/68 (2006.01)
  • A61K 31/70 (2006.01)
  • A61K 38/17 (2006.01)
  • A61K 48/00 (2006.01)
  • C07H 21/04 (2006.01)
  • C07K 14/47 (2006.01)
  • G01N 33/50 (2006.01)
  • G01N 33/574 (2006.01)
  • G01N 33/68 (2006.01)
(72) Inventors :
  • VOGELSTEIN, BERT (United States of America)
  • KINZLER, KENNETH W. (United States of America)
  • SHERMAN, MICHAEL I. (United States of America)
(73) Owners :
  • THE JOHNS HOPKINS UNIVERSITY (United States of America)
  • PHARMAGENICS, INC. (United States of America)
(71) Applicants :
  • THE JOHNS HOPKINS UNIVERSITY (United States of America)
  • PHARMAGENICS, INC. (United States of America)
(74) Agent: SIM & MCBURNEY
(74) Associate agent:
(45) Issued: 2006-01-03
(22) Filed Date: 1992-06-10
(41) Open to Public Inspection: 1992-12-15
Examination requested: 1999-05-28
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): No

(30) Application Priority Data:
Application No. Country/Territory Date
715,182 United States of America 1991-06-14
860,758 United States of America 1992-03-31

Abstracts

English Abstract



Specific sequences in the human genome are the sites of strong
binding of wild-type p53 protein, but not mutant forms of the protein.
These sequences are used diagnostically to detect cells in which the
amount of wild-type p53 is diminished. The sequences can also be used
to screen for agents which correct for loss of wild-type p53 to DNA in
cancer cells.


Claims

Note: Claims are shown in the official language in which they were submitted.



-49-


CLAIMS

1. A method for detecting the presence of wild-type p53 protein in a cell,
comprising the steps of:
contacting a p53-specific binding DNA fragment which comprises more than
one monomer of the sequence 5'-RRRCWWGYYY-3' with a cell lysate from a tissue
of a human, to bind the DNA fragment to wild-type p53 present in the cell
lysate;
detecting the presence of wild-type p53 protein in the cell by detecting
binding
of the DNA fragment to wild-type p53.
2. The method of claim I wherein the p53-specific binding DNA segment
comprises nucleotides 103 to 134 as shown in SEQ ID No: 1.
3. The method of claim 1 wherein the p53-specific binding DNA segment
comprises nucleotides 104 to 123 as shown in SEQ ID No:1.
4. The method of claim 1 wherein there are from 0 to 40 nucleotides between
said monomers.
5. The method of claim 1 wherein the DNA fragment is labelled with a
detectable moiety selected from the group consisting of: a radioactive moiety,
a
colorimetric moiety, and a fluorescent moiety.
6. The method of claim 1 wherein the step of determining the amount of p53-
specific binding DNA comprises:
immunoprecipitating p53 protein with anti-p53 monoclonal antibodies.
7. A method of detecting the presence of a wild-type p53 protein in a cell,
comprising the steps of:
providing a histological section from a human;
incubating the section with a detectably-labeled p53-specific binding DNA
fragment which comprises more than one monomer of the sequence


-50-


5'-RRRCWWGYYY-3' to bind said DNA fragment to wild-type p53 present in the
histological sample;
removing unbound DNA fragment from the histological section; and
determining the amount of DNA fragment which is bound to the histological
sample.
8 The method of claim 7 wherein the p53-specific binding DNA segment
comprises nucleotides 103 to 134 as shown in SEQ ID NO:1.
9. The method of claim 7 wherein the p53-specific binding DNA segment
comprises nucleotides 104 to 123 as shown in SEQ ID NO:1.
10. The method of claim 7 wherein there are between 0 and 40 nucleotides
between said monomers.
11. The method of claim 7 wherein the DNA fragment is labelled with a
detectable moiety selected from the group consisting of: a radioactive moiety,
a
colorimetric moiety, and a fluorescent moiety.
12. The use of a compound to provide the physiological effect of wild-type p53
protein to a cell, wherein the compound comprises a single-stranded, linear or
circular, oligonucleotide or oligonucleotide containing nucleotide analogs
which can
form a complex with a p53 specific DNA binding site, wherein said
oligonucleotide
containing nucleotide analogs comprises more than one monomer of the sequence
5'-RRRCWWGYYY-3'.
13. The use of claim 12 wherein the compound comprises sequences which are
adjacent to said monomer sequence in the human genome.
14. The use of claim 12 wherein the oligonucleotide or oligonucleotide
containing
nucleotide analogs comprises between 0 and 40 nucleotides between said
monomers.


-51-


15. A double-stranded DNA fragment which comprises a p53-specific DNA
binding site, wherein the fragment comprises more than one monomer of the
sequence
RRRCWWGYYY and wherein the fragment is covalently attached to an insoluble
polymeric support.
16. A linear or circular single-stranded oligonucleotide or oligonucleotide
containing nucleotide analogs which is able to complex specifically with a p53-

specific binding site, said binding site comprising more than one monomer of
the
sequence RRRCWWGYYY.
17. The oligonucleotide or oligonucleotide containing nucleotide analogs of
claim
16 which contains one or more switchback linkers which allow the
oligonucleotide to
complex with both strands of the p53-specific binding site.
18. The oligonucleotide or oligonucleotide containing nucleotide analogs of
claim
16 which is selected from the group consisting of a methylphosphonate, an
aminomethylphosphonate, a phosphorothioate, a phosphorodithioate, a
substituted or
unsubstituted phosphoramidate, an oligoribonucleotide, an oligodeoxyribonucleo
tide,
an alpha-oligonucleotide and mixtures thereof.
19. The oligonucleotide or oligonucleotide containing nucleotide analogs of
claim
16 which is terminated at the 3' or 5' end with a moiety which reduces
susceptibility
to oligonucleotide degradation or facilitates uptake by the cells.
20. The oligonucleotide or oligonucleotide containing nucleotide analogs of
claim
19 wherein the moiety is selected from the group consisting of: a substituted
or
unsubstituted amino moiety, polyethylene glycol, polylysine, acridine,
dodecanol, and
cholesterol.
21. A method of identifying compounds which specifically bind to p53-specific
DNA binding sequences, comprising the steps of:


-52-


contacting a p53-specific-binding DNA fragment comprising more than one
monomer of the sequence RRRCWWGYYY with a test compound to bind the test
compound to the DNA fragment;
determining the amount of test compound which is bound to the DNA
fragment.
22. The method of claim 21 wherein soluble DNA fragments are incubated with
the test compound and the p53-specific-binding DNA fragment immobilized on a
solid support, said soluble DNA fragments not having the ability to
specifically bind
wild-type p53 protein.
23. A method of identifying compounds which specifically bind to p53-specific
DNA binding sequences, comprising the steps of:
contacting a p53-specific-binding DNA fragment comprising more than one
monomer of the sequence RRRCWWGYYY immobilized on a solid support with
both a test compound and wild-type p53 protein to bind the wild-type p53
protein to
the DNA fragment;
determining the amount of wild-type, p53 protein which is bound to the DNA
fragment, inhibition of binding of wild-type p53 protein by the test compound
indicating binding of the test compound to the p53-specific DNA binding
sequences.
24. A method of pre-screening agents for use in cancer therapy, comprising:
measuring the amount of binding of a p53 protein encoded
by a mutant gene found in cancer cells of a patient to a double-stranded DNA
molecule which conforms to the consensus binding site having more than one
monomer of RRRCWWGYYY;
measuring the amount of binding of said p53 protein to said DNA molecule in
the presence of a test substance; and
comparing the amount of binding of the p53 protein in the presence of said
test
substance to the amount of binding of the p53 protein in the absence of said
test
substance, said test substance which increases the amount of binding being a
candidate for use in cancer therapy.


-53-


25. A method of pre-screening agents for use in cancer therapy, comprising:
contacting a transfected cell with a test substance, said transfected cell
containing a
p53 protein which is encoded by a mutant gene found in cancer cells of a
patient and a
reporter gene construct comprising a reporter gene which encodes an assayable
product and a double-stranded sequence which conforms to the p53 consensus
binding site having more than one monomer of RRRCWWGYYY, wherein said
sequence is upstream from and adjacent to said reporter gene; and
determining whether the amount of expression of said reporter gene is altered
by the test substance, said test substance which alters the amount of
expression of said
reporter gene being a candidate for use in cancer therapy.
26. A method of pre-screening agents for use in cancer therapy, comprising:
adding RNA polymerase and ribonucleotides to a transcription construct, said
transcription construct comprising a reporter gene which encodes an assayable
product and a double-stranded sequence which conforms to the p53 consensus
binding site having at least two monomers of RRRCWWGYYY, said sequence being
upstream from and adjacent to said reporter gene, said step of adding being
effected in
the presence and absence of a test substance; and determining whether the
amount of
transcription of said reporter gene is altered by the presence of said test
substance, a
test substance which alters the amount of transcription of said reporter gene
being a
candidate for use in cancer therapy.
27. A DNA construct for use in screening potential chemotherapeutic agents,
comprising:
a reporter gene which encodes an assayable product;
a sequence which conforms to the p53 consensus binding
site having more than one monomer of RRRCWWGYYY upstream from and adjacent
to said reporter gene, wherein said DNA construct is selected from the group
consisting of a recombinant plasmid, a viral vector and an isolated molecule
of DNA.
28. A method of diagnosing tumor-inducing or hyperplastic-inducing strains of
human papilloma virus (HPV) comprising:



-54-

contacting cells or cell extracts of patients suspected of being infected by
HPV
with a p53-specific binding DNA fragment comprising more than one monomer of
the
sequence RRRCWWGYYY;
detecting the amount of wild-type p53 in said cells or cell extract which
binds
to said DNA fragment, absence of bound p53 indicating infection by strains of
HPV
which sequester p53.

29. ~A method of pre-screening agents for use in cancer therapy, comprising:
contacting a transfected cell with a test substance, said transfected cell
containing a wild-type p53 protein and a reporter gene construct comprising a
reporter
gene which encodes an assayable product and a sequence which conforms to the
p53
consensus binding site having more than one monomer of RRRCWWGYYY, wherein
said sequence is upstream from and adjacent to said reporter gene; and
determining whether the amount of expression of said reporter gene is altered
by the test substance, a test substance which alters the amount of expression
of said
reporter gene being a candidate for use in cancer therapy.

30. ~A method of pre-screening oligonucleotides for use in cancer therapy,
comprising:
adding a p53 protein which is encoded by a mutant gene found in a cancer
patient and a preparation of random oligonucleotides to a p53-specific-binding
DNA
fragment comprising more than one monomer of the sequence RRRCWWGYYY
immobilized on a solid support;
recovering the oligonucleotides which bound to the solid support.

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02070979 2003-07-03
SEQUENCE SPECIFIC DNA BINDING BY P53
TECHNICAL FIELD OF THE INVENTION
This invention relates to diagnostic and therapeutic methods for
detecting and alleviating cancers. Specifically it relates, to the
identification
of DNA sequences specifically bound by tumor suppresser p53.
BACKGROUND OF THE INVENTION
The gene for the nuclear phosphoprotein p53 is the most commonly
mutated gene yet identified in human cancers (Vogelstein, B., Nature,
348:681 ( 1990)). Missense mutations occur in tumors of the colon, lung,
breast, ovary, bladder, and several other organs (S.J. Baker, et al., Science,
244:217 (1989); J.M. Nigro, et al., Nature, 342:705 (1989); T. Takahashi, et
al., Science, 246:491 (1989); Romano, et al., Oncog~ene, 4:1483 (1989),
Menon, Proc. Natl Acad. Sci.. USA, 87:5435 (1990); Iggo, et al., Lancet ii,
675 ( 1990); T. Takahashi, et al., J. Clin. Invest. 86:363 ( 1990); Mulligan,
Proc. Natl Acad. Sci. USA, 87:5863 (1990); Bartek, et al., Onco~ene, 5:893
( 1990); Stratton et al., Oncog_ene, 5:1297 ( 1990)). One of the important
challenges of current cancer research is the elucidation of the biochemical
properties of the p53 gene product and the way in which mutations of the
p53 gene affect these properties.
Although some biological characteristics of p53 have been defined,
such as its ability to suppress the growth of in vitro transformed murine
cells
(Eliyahu, et al, Proc. Natl Acad. Sci. USA 86:8763 (1989); Finlay, et al.,
Cell, 57:1083 ( 1989)) or human cancer cells (Baker, et al., Science, 249:912
( 1990); Mercer, et al., Proc. Natl



r ~ ~ ~ 20709?9
-2-
Acad. Sci. USA, 87:6166 (1990); Diller et al., Mol. Cell Biol. 10:5772
(1990)), the biochemical basis of this suppression remains largely
unknown. As a step towards understanding such properties, we have
attempted to determine whether p53 binds to specific DNA sequences
within the human genome.
SUMMARY OF THE INVENTION
It is an object of the invention to provide a method for detecting
the presence of wild-type p53 protein in a cell.
It is another object of the invention to provide a method for
providing the physiological effect of wild-type p53 protein to a cell.
It is yet another object of the invention to provide a
double-stranded DNA fragment which contains a p53-specific DNA
binding site.
It is yet another object of the invention to provide a
single-stranded oligonucleotide or oligonucleotide containing nucleotide
analogs which can specifically complex to a p53-specific DNA binding
site.
It is still another object of the invention to provide methods for
identifying compounds which specifically bind to p53-specific DNA
binding sequences.
It is still another object of the invention to provide methods for
identifying compounds which restore the ability of mutant p53 proteins
to bind to, Specific DNA binding sequences.
These and other objects of the invention are provided by one or
more of the embodiments described below. In one embodiment a
method is provided for detecting the presence of wild-type p53 protein
in a cell, comprising the steps of: contacting a p53-specific-binding
DNA fragment with a cell lysate from a tissue of a human to bind the
DNA fragment to wild-type p53 present in the cell lysate; and
detecting the binding of the p53-specific-binding DNA fragment to
wild-type p53.
In another embodiment of the invention a method is disclosed
for providing the physiological effect of wild-type p53 protein to cell,
comprising the steps of: providing to a cell a compound which is able
to specifically complex with a p53-specific binding site.




2070979
-3-
In yet another embodiment a double-stranded DNA fragment is
provided which comprises a p53-specific-DNA binding site, wherein the
fragment comprises more than one monomer repeat of the sequence
5'-RRRCWWGYYY-3~ and wherein the fragment is covalently attached
to an insoluble polymeric support.
In another embodiment of the invention a single-stranded
oligonucleotide containing natural nucleotides and/or nucleotide
analogs is provided which is able to complex specifically with a
p53-specific binding site, said binding site comprising more than one
monomer of the sequence 5'-RRRCWWGYYY-3'.
In yet another embodiment of the invention a method is provided
for identifying compounds which specifically bind to p53-specific DNA
binding sequences, comprising the steps of: contacting a p53-specific
DNA binding fragment immobilized on a solid support with a test
compound to bind the test compound to the DNA fragment; and
determining the amount of test compound which is bound to the DNA
fragment.
In even another embodiment of the invention a method is
provided for identifying compounds which specifically bind to
p53-specific-DNA binding sequences, comprising the steps of:
contacting a p53-binding DNA fragment immobilized on a solid support
with both a test compound and wild-type p53 protein to bind the
wild-type p53 protein to the DNA fragment; determining the amount of
wild-type p53 protein which is bound to the DNA fragment, inhibition
of binding of wild-type p53 protein by the test compound suggesting
binding of the test compound to the p53-specific DNA binding
sequences.
In still another embodiment a method of prescreening agents for
use in cancer therapy is provided comprising: measuring the amount of
binding of a p53 protein which is encoded by a mutant gene found in
cancer cehs of a patient to a DNA molecule which comprises more
than one monomer of RRRCWWGYYY; measuring the amount of
binding of said p53 protein to said DNA molecule in the prESence of a
test substance; and comparing the amount of binding of the p53 protein
in the presence of said test substance to the amount of binding of the




. . _ 4 _ 2070979
p53 protein in the absence of said test substance, a test substance
which increases the amount of binding being a candidate for use in
cancer therapy.
In another embodiment of the invention a method is provided for
prescreening agents for use in cancer therapy comprising: contacting
a transfected cell with a test substance, said transfected cell
containing a p53 protein which is encoded by a mutant gene found in
cancer cells of a patient and a reporter gene construct comprising a
reporter gene which encodes an assayable product and a sequence
which conforms to the p53 consensus binding site having more than one
monomer of RRRCWWGYYY, wherein said sequence is upstream from
and adjacent to said reporter gene; and determining whether the
amount of expression of said reporter gene is altered by the test
substance, a test substance which alters the amount of expression of
said reporter gene being a candidate for use in cancer therapy.
In still another embodiment a method of prescreening agents for
use in cancer therapy is provided comprising: adding RNA polymerase
and ribonucleotides to a transcription construct, said transcription
construct comprising a reporter gene which encodes an assayable
product and a sequence which conforms to the p53 consensus binding
site having more than one monomer of RRRCWWGYYY, said sequence
being upstream from and adjacent to said reporter gene, said step of
adding being effected in the presence and absence of a test substance;
determining whether the amount of transcription of said reporter gene
is altered by the presence of said test substance, a test substance which
alters the amount of transcription of said reporter gene being a
candidate for use in cancer therapy.
In a further embodiment a DNA construct is provided
comprising: a reporter gene which encodes an assayable product; and a
sequence which conforms to the p53 consensus binding site having
more than one monomer of RRRCWWGYYY upstream from and
adjacent to said reporter gene; wherein said DNA construct is selected
from the group consisting of a recombinant plasmid, a viral vector or
an isolated molecule of DNA.

CA 02070979 2003-07-03
-5-
In another embodiment of the invention a method is provided of diagnosing
tumor-inducing or hyperplastia-inducing strains of human papillioma virus
(HPV)
4 comprising: contacting cells or cell extracts of patients suspected of being
infected by
HPV with a p53-specific binding DNA fragment; and detecting the amount of wild-

type p53 in said cells or cell extract which binds to said DNA fragment,
absence of
bound p53 indicating infection by strains of HPV which sequester p53.
8 In accordance with an aspect of the invention, a method for detecting the
presence of wild-type p53 protein in a cell, comprising the steps of:
contacting a p53-specific binding DNA fragment which comprises more than
one monomer of the sequence 5'-RRRCWWGYYY-3' with a cell lysate from a tissue
12 of a human, to bind the DNA fragment to wild-type p53 present in the cell
lysate;
detecting the presence of wild-type p53 protein in the cell by detecting
binding
of the DNA fragment to wild-type p53.
In accordance with a further aspect of the invention, a method of detecting
the
16 presence of a wild-type p53 protein in a cell, comprising the steps of:
providing a histological section from a human;
incubating the section with a detectably-labeled p53-specific binding DNA
fragment which comprises more than one monomer of the sequence 5'-
20 RRRCWWGYYY-3' to bind said DNA fragment to wild-type p53 present in the
histological sample;
removing unbound DNA fragment from the histological section; and
determining the amount of DNA fragment which is bound to the histological
24 sample.
In accordance with a another aspect of the invention, there is provided
the use of a compound to provide the physiological effect of wild-type p53
protein to
a cell, wherein the compound comprises a single-stranded, linear or circular,
28 oligonucleotide or oligonucleotide containing nucleotide analogs which can
form a
complex with a p53 specific DNA binding site, wherein said oligonucleotide
containing nucleotide analogs comprises more than one monomer of the sequence
5'-
RRRCWWGYYY-3'.


CA 02070979 2003-07-03
-5 a-
In accordance with a further aspect of the invention, a double-stranded DNA
fragment which comprises a p53-specific DNA binding site, wherein the fragment
4 comprises more than one monomer of the sequence RRRCWWGYYY and wherein
the fragment is covalently attached to an insoluble polymeric support.
In accordance with another aspect of the invention, a linear or circular
single-
stranded oligonucleotide or oligonucleotide containing nucleotide analogs
which is
8 able to complex specifically with a p53-specific binding site, said binding
site
comprising more than one monomer of the sequence RRRCWWGYYY.
In accordance with a further aspect of the invention, a method of identifying
compounds which specifically bind to p53-specific DNA binding sequences,
12 comprising the steps of:
contacting a p53-specific-binding DNA fragment comprising more than one
monomer of the sequence RRRCWWGYYY with a test compound to bind the test
compound to the DNA fragment;
16 determining the amount of test compound which is bound to the DNA
fragment.
In accordance with a yet a further aspect of the invention, a method of
identifying compounds which specifically bind to p53-specific DNA binding
20 sequences, comprising the steps of:
contacting a p53-specific-binding DNA fragment immobilized on a solid
support with both a test compound and wild-type p53 protein to bind the wild-
type
p53 protein to the DNA fragment;
24 determining the amount of wild-type p53 protein which is bound to the DNA
fragment, inhibition of binding of wild-type p53 protein by the test compound
indicating binding of the test compound to the p53-specific DNA binding
sequences.
In accordance with a further aspect of the invention, a method of pre-
screening
28 agents for use in cancer therapy, comprising:
measuring the amount of binding of a p53 protein encoded by a mutant gene
found in cancer cells of a patient to a double-stranded DNA molecule which
conforms
to the consensus binding site having more than one monomer of RRRCWWGYYY;
32

CA 02070979 2003-07-03
-Sb-
measuring the amount of binding of said p53 protein to said DNA molecule in
the presence of a test substance; and
4 comparing the amount of binding of the p53 protein in the presence of said
test
substance to the amount of binding of the p53 protein in the absence of said
test
substance, a test substance which increases the amount of binding being a
candidate
for use in cancer therapy.
8 In accordance a further aspect of the invention, a method of pre-screening
agents for use in cancer therapy, comprising:
contacting a transfected cell with a test substance, said transfected cell
containing a p53 protein which is encoded by a mutant gene found in cancer
cells of a
12 patient and reporter gene construct comprising a reporter gene which
encodes an
assayable product and a double-stranded sequence which conforms to the p53
consensus binding site having more than one monomer of RRRCWWGYYY, wherein
said sequence is upstream from and adjacent to said reporter gene; and
16 determining whether the amount of expression of said reporter gene is
altered
by the test substance, a test substance which alters the amount of expression
of said
reporter gene being a candidate for use in cancer therapy.
In accordance with yet a further aspect of the invention, a method of pre-
20 screening agents for use in cancer therapy, comprising:
adding RNA polymerase and ribonucleotides to a transcription construct, said
transcription construct comprising a reporter gene which encodes an assayable
product
and a double-stranded sequence which conforms to the p53 consensus binding
site
24 having at least two monomers of RRRCWWGYYY, said sequence being upstream
from and adjacent to said reporter gene, said step of adding being effected in
the
presence and absence of a test substance; and
determining whether the amount of transcription of said reporter gene is
28 altered by the presence of said test substance, a test substance which
alters the amount
of transcription of said reporter gene being a candidate for use in cancer
therapy.
According to a further aspect of the invention, a DNA construct for use in
screening potential chemotherapeutic agents, comprising:
32 a reporter gene which encodes an assayable product;


CA 02070979 2004-12-17
SC
a sequence which conforms to the p53 consensus binding site having more than
one monomer of RRRCWWGYYY upstream from and adjacent to said reporter gene,
wherein said DNA construct is selected from the group consisting of a
recombinant
plasmid, a viral vector and an isolated molecule of DNA.
According to a further aspect of the invention, a method of diagnosing tumor-
inducing or hyperplastic-inducing strains of human papilloma virus (HPV)
comprising:
contacting cells or cell extracts of patients suspected of being infected by
HPV
with a p53-specific binding DNA fragment comprising more than one monomer of
the
sequence RRRCWWGYYY;
detecting the amount of wild-type p53 in said cells or cell extract which
binds to
said DNA fragment, absence of bound p53 indicating infection by strains of HPV
which
sequester p53.
According to a further aspect of the invention, a method of pre-screening
agents
for use in cancer therapy, comprising:
1 S contacting a transfected cell with a test substance, said transfected cell
containing
a wild-type p53 protein and a reporter gene construct comprising a reporter
gene which
encodes an assayble product and a sequence which conforms to the p53 consensus
binding site having more than one monomer of RRRCWWGYYY, wherein said sequence
is upstream from and adjacent to said reporter gene; and
determining whether the amount of expression of said reporter gene is altered
by
the test substance, a test substance which alters the amount of expression of
said reporter
gene being a candidate for use in cancer therapy.
According to a further aspect of the invention, a method of pre-screening
oligonucleotides for use in cancer therapy, comprising:
adding a p53 protein which is encoded by a mutant gene found in a cancer
patient
and a preparation of random oligonucleotides to a p53-specific-binding DNA
fragment
comprising more than one monomer of the sequence RRRCWWGYYY immobilized on a
solid support;
recovering the oligonucleotides which bound to the solid support.
According to a further aspect of the invention, a method of identifying
compounds which specifically bind to p53-specific DNA binding sequences,
comprising
the steps of
contacting a p53-specific-binding DNA fragment comprising more than one


' CA 02070979 2004-12-17
Sd
monomer of the sequence RRRCWWGYYY immobilized on a solid support with both a
test compound and wild-type p53 protein to bind the wild-type p53 protein to
the DNA
fragment;
determining the amount of wild-type, p53 protein which is bound to the DNA
fragment, inhibition of binding of wild-type p53 protein by the test compound
indicating
binding of the test compound to the p53-specific DNA binding sequences.
These and other embodiments of the invention provide the art with new tools
for
detecting and remedying the loss of function of the most commonly mutated gene
identified in human cancers. Further it provides methods for identifying
agents which
can suppress neoplastic growth.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure lA. Screening for fragments bound by p53 using an immunoprecipitation
assay. Panel 1 contains the hFosAva2 clone; panel 2, 772 CBE; panel 3, Lambda
SR;
panel 4, a pool of clones with inserts of randomly cloned human genomic
sequences. 772
CBE and Lambda SR contain HinfI fragments (259 and 190 bp, respectively) which
bound p53 relatively strongly (arrowheads). "C" - control lane, containing 2%
of the
labelled DNA used in the binding reactions. "B"- bound DNA recovered from the
immunoprecipitate. Figure 1B. Tests for dependence on p53 and specific
antibody. Cell
lysates were produced by infection with vaccinia virus that did (+) or did not
(-) contain
an insert of wild-type p53 cDNA. Immunoprecipitation was performed with anti-
p53
monoclonal antibodies (+) or normal mouse IgG (-).
Figure 2. Relative abilities of wild-type and mutant p53 to precipitate
fragment
A. "C" - control lanes, containing 2% of the labelled DNA used in the binding
reaction,
"B" - bound DNA recovered from the immunoprecipitate. Figure 2A. Increasing
quantities of wild-type and mutant 273his p53, affinity-purified from a
baculovirus
expression system, were used to precipitate labelled CBE fragments. Figure 2B.
Lysates
from a vaccinia virus system (Vac) producing the wild-type (wt), mutant
(175his), or no
p53 protein (-), were used to immunoprecipitate labelled CBE fragments.
Equivalent
quantities of



2070979
-6-
p53 were present in the wild-type and mutant p53 lysates, as assessed
by Western blot. In the "Bac" lane, affinity-purified p53 produced in
baculovirus-infected insect cells was used in place of the
vaccinia-infected lysates.
Figure 3A. Fragment A and adjacent vector sequences in the
lOd subclone of 772 CHI. This differs from the published 772 CHE
sequence (Genbank M25718) in the number of CTT repeats (bp 173-229)
and in the presence of A instead of C at by 116. Figure 3B. Fragment
B and adjacent vector sequences in the 8a subclone of Lambda 5R. A
related sequence on file (Genbank X05913) varied somewhat from the
Lambda SR subclone studied.
Figure 4. Binding of various subfragments of fragments A and B
to p53 from vaccinia-infected cell lysates. Figure 4A. Subfragments of
fragment A (subclone lOd) were assayed by immunoprecipitation for
their ability to bind wild-type p53 from vaccinia-infected cell lysates.
Binding of at least 296 of the DNA added to the reaction was judged as a
positive (+) result; lesser but significant binding was recorded as "+/-".
Double Lines (_) denote fragment A sequences. Single lines (-) denote
polylinker sequences of the vector, not originally present in fragment
A (Fig. 1). Fragment 5mut1 had a G to T transversion at by 120; 5mut2
had G to T transversions at by 120 to 122. Figure 4B. The fragment A
(panels 1-4) and fragment B (panel 5) subfragments illustrated in Figure
4A are labelled to the lef t of the bands. The 'w't band in panel 4
corresponds to the 2.9 kb vector into which subfragment 6 was cloned.
Subfragment 8 (panel 5) contained by 104-238 of fragment B (see Fig.
3B). Control lanes (C) contained 296 of the labelled fragments used in
the binding assays (B).
Figure 5. Effects of methylation and point mutations in
fragment A on DNA-binding. Figure 5A. Methylation interference
assay. "B'~ - bound DNA recovered from the immunoprecipitate. "C" -
equivalent amount of control DNA fragments, not subject to binding
reaction. Bound and control DNA samples were cleaved at methylated
guanines and equal amounts separated by electrophoresis on a 696
denaturing gel. Dots represent methylation-sensitive sites (open for
partial, solid for strong interference); some variation in band




20?4979
_?_
intensities occurred between assays, and only the reproducible changes
are marked. Figure 5B. Binding of "mutant" subfragments of fragment
A (5mut1 and 5mut2) to purified baculovirus-produced p53 is compared
to that of the normal subfragment 5 sequence. 5mut1 contains a T
instead of G at by 120, and 5mut2 contains Ts in place of Gs at by 120,
121, and 122.
Figure 6A shows RNase protection analysis of transfected clonal
lines. A labeled antisense p53 probe was hybridized with total cellular
RNA, and digested with RNase A. Endogenous RNA included all
sequences represented in the labeled probe. Exogenous p53 RNA
produced from the expression vectors extended only about 2/3 of the
length of the probe.
Figure 6B shows Southern blot analysis of transfected clonal
lines. The exogenous p53 gene was present on a 1.8 kb BamHI
fragment. The endogenous p53 gene gave rise to a ?.8 kb BamHI
fragment. Other sized fragments presumably arose by rearrangements.
Figure ?A shows expression analysis of pooled clones; the
analysis was as described in Figure 6A.
Figure ?B shows a Southern blot analysis of SW480 pooled clones.
Figure 8. Isolation of human genomic sequences which bound to
p53.
Figure 8A. Experimental strategy used for isolation and analysis
of human genomic DNA fragments which bound to p53.
Figure 8B. Immunoprecipitation (IP) assays of cloned fragments.
Clones of amplified and selected (AS) DNA were tested for the
presence of p53-binding fragments by IP. For each clone, the bound
DNA is shown in the B lane, adjacent to a control (c) lane containing
296 of the total end-labeled DNA used in the binding assay. In this
representative experiment, eight binding fragments were identified,
representing six unique genomic fragments. The inserts from the
clones in lanes labeled, 2, 3, 5, 9, 10, and 11 contained p53-binding
fragments, while the other lanes contained none. The clones in lanes 2
and 5 each contained two binding fragments.
Figure 9. Mapping of p53-binding sites by DNase I protection
(DP) and methylation interference (MI). For each footprint, the first

CA 02070979 2003-07-03
- g -
and fourth lanes contain control samples of the total labeled DNA, whereas the
middle
two lanes contain an equivalent amount of p53-bound DNA. DNA sequences
corresponding to the p53-binding regions on strand 1 are shown in Fig. 10.
Figure 10. Definition of a consensus binding site for p53. The p53 binding
site of 18 cloned human genomic DNA fragments, determined by footprinting
methods are displayed along the central axis of symmetry which separates the
two 10
by consensus monomers. Nucleotides in capital letters represent identity of a
genomic sequence to the consensus, whereas lower case letters identify
disparity with
the consensus. Sequences surrounding the consensus or separating the two 10 by
monomers are also shown in lower case. The ten synthetic oligonucleotides
investigated for the ability to be bound by p53 are shown at the bottom.
Oligonucleotides No. 6 to 10 were tested after cloning into plasmid vectors.
Lower
case letters represent vector-derived sequences. Combined nucleotide usage (%)
within the two monomers of the consensus binding site is shown in the middle.
Figure 11. Binding of synthetic oligonucleotides to wild-type (wt) and mutant
p53 proteins.
Figure 11A. The 10 by consensus monomer was insufficient for binding,
whereas dimers in various orientations or multimers of the 10 by consensus
bound
strongly to p53. For each sample, the control lane (C) contained 2% of the
total DNA
used in the binding reaction, and was composed of two fragments: 2.9 kb vector
DNA
fragment, and a fragment of 40 to 80 by containing no insert (lane 9; XhoI
plus Pstl
digest of pBluescript T"" II SK+), the 10 by consensus monomer sequence 5'-
AGGCATGTCT-3' (lane 5), or multimers of this sequence arranged as indicated
(lanes 1 to 4, 6, to 8). Bound DNA from the IP is shown in the B lanes.
Figure 11B. Comparison of the ability of wild-type and mutant p53 to bind to
the consensus dimer. In vitro translated p53 proteins were tested for the
ability to
bind the consensus dimer by IP. Two percent of the total DNA used for binding
is
shown in lane 1. Lane 7 shows binding to baculovirus-produced human wild-type
p53 protein. Lanes 2 to 6 show binding of in vitro translated wild-type and
mutant
p53 proteins. The mutant p53 proteins contained changes at codon 143 (val to
ala),


CA 02070979 2003-07-03
-9-
175 (arg to his), 248 (arg to trp), and 273 (arg to his).
Figure 12. Reporter and expression constructs used in transfections are
shown.
Figure 12A. Reporter constructs in pBluescript II SK + vector (Stratagene).
PG", concatemers of n copies of the p53 binding sequence PG. MG", concatemers
of
the mutated sequence, which does not bind p53. CAT, the chloramphenicol
acetyltransferase coding sequence. LacZ, the (3-galactosidase coding sequence.
Py,
the early gene promoter from polyomavirus. CYC, the yeast cytochrome c gene
promoter.
Figure 12B. Expression vectors. CMV, the cytomegalovirus promoter from
the parent vector pCMVneoBam. Gal, the galactose-inducible promoter from
yeast.
Figure 13. Correlation of DNA-binding and transactivation is shown.
Figure 13A. Relative DNA-binding abilities of various length concatemers of
a p53-binding sequence (PG" series), using an immunoprecipitation assay.
Clones
were cleaved by restriction endonucleases to extricate the concatemers, end-
labelled,
incubated with purified baculovirus-produced wild-type human p53,
immunoprecipitated with anti-p53 and protein A-Sepharose T"~1 and bound
fragments
recovered and separated on a nondenaturing polyacrylamide gel. C, control
lane,
containing 2% of the labeled DNA used in the binding reactions. B, bound DNA
recovered from the binding reactions.
Figure 13B. Transactivation efficiencies of reporters containing the various
PG" concatemers compared by chloramphenicol acetyltransferase (CAT) assay. 1.7
~,g of the expression vector p53-wt were transfected into HCT 116 cells.
Reporters
had one orientation of the PG" sequence (...TGCCT...Py...CAT...), except for
PGZ-
CAT and PG,3-CAT, which had the opposite orientation (...AGGCA...Py...CAT...).
Results are expressed relative to the CAT activity in lane 7, which was
arbitrarily set
at 100.
Figure 14. Comparison of the ability of wild-type and mutant p53 to bind to a
concatemerized binding sequence (PG,6), and lack of




2070979
-lU-
DNA-binding ability to MG15 are shown. Each p53 form (as a rabbit
reticulocyte preparation (Promega) made essentially as described in
Kern, et al., Oncor~ene, 6:131-136 (1991); equalized for p53 quantity by
immunoblot analysis, data not shown) was used to immunoprecipitate
end-labeled DNA. C, control lanes, containing 296 of the labeled DNA
used in the binding reaction. B, bound DNA recovered from the
immunoprecipitate.
Figure 15. Effects of varying the location of the binding
sequence relative to the promoter are shown.
Figure 15A. Efficiencies of reporters with PG13 separated by
varying distances from the polyomavirus promoter. Various lengths of
the non-binding sequence (MG" series) were used to provide inert
spacers of defined length (see Fig. 8A). CAT assays are shown.
Figure 15B. Transactivation efficiency of a reporter with PGIs
downstream of CAT (CAT-PG16). 1.7 ug each of expression and
reporter construct were transfected. The MG15-CAT served as a
negative control. CAT assays are shown.
Figure 16. Relative abilities of wild-type and mutant p53 to
activate transcription are shown.
Figure 16A. Representative CAT assays (Exp. 2 from Table 1).
Figure 16B. Western blot analysis of p53 expression in lysates of
transfected cells, showing levels of expression at least as high as that
of wild-type p53 were obtained with the mutant p53 clones. In one
case (143a1a), the level of expression of p53 (lane 3) was slightly lower
than with the other constructs, for unknown reasons. When 2.55 ug of
vector was used in the transfection, however, the level produced by the
p53-143 vector (lane Z) was at least as high as that for the p53-wt
vector, but no transactivation was observed (Table I).
Figure 1?. Effect of co-expressed 1'15''ig mutant p53 on
transactivation by wild-type p53 is shown. 1.? ug of PG13-CAT
reporter was used with the stated amounts of p53 expression
constructs. The balance of transfected plasmid was pCMVneoBam (for
expressor) and pBluescript II SK+ (for reporter) to a total of 5.1 ug.
CAT assays are shown.




v 2070979
-11-
Figure 18. Dominant-negative effects of various mutant p53
proteins are demonstrated. 0.85 ug of p53-wt was used in all
transfections, without or with the addition of 0.85 ug or 2.55 ug mutant
p53 construct, or with an additional 2.55 ug p53-wt. 1.? ug PG13-CAT
reporter was used in each case. The composite results shown are
representative of at least two transfections done on separate days.
Figure 19. A biochemical model for the effect of p53 mutations
is depicted. Cellular p53 exists as oligomers. Oligomeric p53 binds its
recognition sequence of DNA (shaded thick line) and activates the
transcription of adjacent genes (filled box). The depiction of p53
dimers is for illustrative purposes only: tetramers and other forms
likely occur. Hetero-oligomerization with mutant proteins inactivates
the transactivation function of the participating wild-type p53
molecules, producing the dominant-negative effect of missense p53
mutations. The remaining activity provided by the residual wild-type
homo-oligomers is lost upon deletion of the remaining wild-type allele,
a frequent event in the progression of human neoplasms.
DETAILED DESCRIPTION OF THE INVENTION
It is a finding of this invention that wild-type p53 protein binds
specific fragments of human chromosomal DNA. Each of the
fragments contains more than one monomer of the double-stranded
motif 5~-RRRCWWGYYY-3~ separated by 0 to 13 bp. Some of these
sequences are found near origins of replication of certain animal
viruses and animal cells. See Jelinek et al, Proc. Natl. Acad. Sci. USA,
vol. ??, pp. 1398-1402 (1980). Four mutant forms of p53 protein which
are commonly Pound in human tumors do not have the ability to bind to
these sequences. Thus, a function of p53 may be mediated by its ability
to bind to specific DNA sequences in the human genome.
Wild-type p53 protein binds specifically to certain DNA
sequences. Two previously isolated DNA fragments, called herein
fragment A and fragment B, have been identified which contain
residues which appear responsible for the binding. These residues are
located within DNA segments which contain nucleotides 103-134 of
SEQ ID NO:1 and nucleotides 104-198 of SEQ ID N0:2.




2070979
-12-
Fragment A contains sequences near a putative replication
origin of the ribosomal gene cluster, while fragment B contains
sequences that may allow adjacent sequences to replicate as
extrachromosomal circles in HeLa cells (Sylvester, et al., Gene 84, 193,
1989). The TGCCT repeat present in the DNA binding region of both of
these fragments has been observed in other potential replication
origins. Thus p53 may be involved in the regulation of initiation of
DNA synthesis by virtue of its binding.
It has been found that p53 will specifically bind to other
sequences in the human genome with similar sequence motifs. Using a
strategy coupling immunoprecipitation to "whole-genome PCR"
(Kinzler, et al., Nucleic Acids Research, i?:3645-3653 (1989)), nineteen
human DNA fragments that bind to p53 have been identified. Each of
the fragments contain a sequence which conforms to a dimer of the
double-stranded motif 5'-RRRCWWGYYY-3~, separated by 0 to 13 bp.
These dimers directly mediate binding, as assessed by DNase I
protection and methylation interference assays. The consensus dimers
contain a striking symmetry, with four 5'-RRRCW-3' units oriented in
alternating directions. A synthetic monomer containing the 10 by
consensus sequence is insufficient for binding, while the combination of
two or more monomers bind strongly to wt p53, but negligibly to p53
mutants. Thus, more than one monomer appears to be required for
binding. The spacing between monomers may be from 0 to 40
nucleotides, although all natural binding sites isolated have spacings of
less than 15 nucleotides. The symmetry of the four half-sites within
the consensus dimers suggests that p53 interacts with DNA as a
tetrameric protein. The eighteen unique clones shown in Figure 10
allow the identification of adjacent genes which may be regulated by
p53 and may mediate its growth-suppressive action.
A sequence which conforms to the consensus sequence need not
have the exact sequence of the dimer. As shown in Figure 10,
p53-binding sequences can have nucleotides which are different than
those designated. There can also be extra nucleotides inserted within
the dimer. Nucleotides can also be missing from the dimer. Typically



2070979
-13-
p53-binding sequences will vary from the consensus sequence at no
more than 5 nucleotide positions.
It is an additional finding of the present invention that wild-type
p53 can activate the expression of genes adjacent to a specific binding
site. Moreover, the level of in vivo transactivation is proportional to
the in vitro strength of DNA-binding. Mutant p53 encoded by
oneogenic p53 genes (i.e., those found in cancer cells of human
patients, completely lose the ability to transactivate. In addition the
mutant p53 proteins exert a dominant-negative effect, dramatically
reducing the transactivating activity of wild-type p53.
Based on the sequence information of the p53 specific-DNA-
binding fragments, a number of diagnostic and therapeutic methods
have been devised. According to one such method, cell lysates are
tested for the presence or absence of wild-type p53 by virtue of its
specific DNA binding ability. As it is known for various cancers and
stages of cancers that one or both of the p53 alleles in tumor tissues
can be mutant, testing for the presence or absence of wild-type p53
protein can provide diagnostic and prognostic information regarding a
tumor and the patient. The cells to be tested are typically isolated
from a tissue suspected of being neoplastic. Preferably the tissues are
carefully prepared and isolated so that non-neoplastic tissues are not
mixed with the neoplastic tissues, which can confound the analysis.
Means for separating neoplastic tissues from non-neoplastic tissues are
known in the art and include dissection of paraffin or cryostat sections,
as well as use of flow cytometry. A cell lysate can be prepared from
the tumor tissue according to any method known in the art. The cell
lysate is then incubated with DNA fragments which are known to bind
the wild-type p53 protein, under conditions which are conducive to
such DNA/protein interactions. Alternatively, a histological sample
can be analyzed by incubation with DNA fragments, as described for
cell lysates.
It is known that p53 also binds non-specifically to DNA. Specific
binding can be distinguished from non-specific binding by any means
known in the art. For example, specific binding interactions are
stronger than non-specific binding interactions. Thus the incubation




2070979
-14-
mixture can be subjected to any agent or condition which destabilizes
protein/DNA interactions such that the specific binding reaction is the
predominant one detected. Alternatively, as taught more specifically
below, a non-specific competitor, such as dI-dC, can be added to the
incubation mixture. If the DNA containing the specific binding sites is
labelled and the competitor is unlabeled, then the specific binding
reactions will be the ones predominantly detected upon measuring
labelled DNA.
According to one embodiment of the invention the DNA which is
bound to p53 is separated from unbound DNA by immunoprecipitation
with antibodies which are specific for p53. Use of two different
monoclonal anti-p53 antibodies may result in more complete
immunoprecipitation than either one alone. Unbound DNA remains in
suspension. The amount of DNA which is in the immunoprecipitate can
be quantitated by any means known in the art. According to one
aspect of the invention, the DNA fragment is labelled with a detectable
moiety, such as a radioactive moiety, a colorimetric moiety or a
fluorescent moiety. Techniques for so labelling DNA are well known in
the art. According to other embodiments of the invention, p53 which
binds to the specific DNA sequence of the present invention can be
detected by gel shif t assays (Tan, Cell, 62:36'1, 1990), nuclease
protection assays (see Example 9, infra), or methylase interference
assays (see Example 9, infra).
According to another embodiment of the invention, after
incubation of p53 with specific binding DNA fragments all components
of the cell lysate which do not bind to the DNA fragments are removed.
This can be accomplished, among other ways, by employing DNA
fragments which are attached to an insoluble polymeric support such as
agarose, cellulose and the like. Af ter binding, all non-binding
components can be washed away, leaving p53 bound to the DNA/solid
support. The p53 can be quantitated by any means known in the art. It
can be determined using an immunological assay, such as an ELISA, RIA
or Western blotting.
The diagnostic assay of the present invention has applicability
not only with regard to cancers which are known to involve mutation




. 2070979
-15-
of p53, .but also with regard to human viruses such as human papilloma
virus (HPV). HPV protein E6 binds tightly to wild-type but not mutant
p53. See Werness et al., Science, 248, ?6-69 (1990). This tight binding
is likely to block the interaction of p53 with its specific DNA binding
sequences. By testing cells or cell extracts suspected of being infected
with potentially tumor-inducing or hyperplastia-inducing strains of
HPV or possibly other viruses, infected cells can be identified, because
the E6 protein of the infected cells will have sequestered the wild-type
p53, rendering it unable to bind to its specific DNA binding sequences.
Such assays may be performed on cell extracts or on histological
specimens.
According to the present invention a method is also provided of
supplying wild-type p53 function to a cell which carries mutant p53
alleles. The wild-type p53 gene or a part of the gene may be
introduced into the cell in a vector such that the gene remains extra-
chromosomal. In such a situation the gene will be expressed by the cell
from the extrachromosomal location. If the mutant p53 genes present
in the cell are expressed, then the wild-type p53 gene or gene portion
should be expressed to a higher level than that of the 'mutant gene.
This is because the mutant forms of the protein are thought to
oligomerize with wild-type forms of the protein. (Eliyahu et al.,
Oncogene, vol. 3, p. 313, 1988.) If a gene portion is introduced and
expressed in a cell carrying a mutant p53 allele, the gene portion
should encode a part of the p53 protein which is required for non-
neoplastic growth of the cell. More preferred is the situation where
the wild-type p53 gene or a part of it is introduced into the mutant cell
in such a way that it recombines with the endogenous mutant p53 gene
present in the cell. Such recombination would require a double
recombination event which would result in the correction of the p53
gene mutation. Vectors for introduction of genes both for
recombination and for extrachromosomal maintenance are known in
the art and any suitable vector may be used.
Polypeptides or other molecules which have p53 activity may be
supplied to cells which carry mutant p53 alleles. The active molecules
can be introduced into the cells by microinjeetion or by liposomes, for




zo7o979
-16-
example. Alternatively, some such active molecules may be taken up
by the cells, actively or by diffusion. Supply of such active molecules
will effect a less aggressive state of neoplasia.
According to the present invention compounds which have p53
activity are those which specifically complex with a p53-specific DNA
binding site. Wild-type p53 is one such compound, but portions of p53
which retain the ability to bind to p53-specific binding sites may also
be used. Oligonucleotides and oligonucleotide containing nucleotide
analogs are also contemplated among those compounds which are able
to complex with a p53-specific DNA binding site. Although applicants
do not wish to be bound by any particular theory, it is believed that
oligonucleotides bind double-stranded DNA to form triplexes. Such
triplexes have been shown to block transcription of certain genes, as
well as protect the DNA binding sites from the action of enzymes such
as DNA methylases. Although originally such oligonucleotides were
thought to require only or predominantly pyrimidines (cytosine and
thymine), purines have also successfully been incorporated into triplex
forming oligonucleotides. Particular oligonucleotides which may be
used include: nucleotides 140-162 of SEQ ID N0:2, nucleotides 128-158
of SEQ ID NO: 1, nucleotides 114-123 of SEQ ID -NO: 1, or portions
thereof having at least ten nucleotides.
Oligonucleotides containing nucleotide analogs or otherwise
modified oligonucleotides may also be useful in the formation of
complexes with double-stranded DNA. For example, certain
modifications to the 3'-terminus can be employed to reduce the
susceptibility of the oligonucleotide to nuclease degradation. Moieties
which may be appended to the 3'- or 5'-termini include a substituted or
unsubstituted amino group, poly~-'~ylene glycol, polylysine, acridine,
dodecanol, and cholesterol. Oligonucleotides and oligonucleotides
containing nucleotide analogs which may be used include
methylphosphonatps, aminomethylphosphonates, phosphorothioates,
phosphorodithioates, substituted or unsubstituted phosphoramidates,
oligoribonucleotides, oligodeoxyribonucleotides, alpha-oligonucleotides
and mixtures thereof. Other modifications to oligonucleotides may be
desirable to increase the uptake by cells or nuclei of the



2070979
-1'I-
oligonucleotides or to decrease nuclease sensitivity. All such
modifications are within the contemplation of the invention.
Switchback linkers may also be incorporated into the midst of an
oligonucleotide or analog. Such linkers are taught by Riordan and
Martin (Nature, 350, 452, 1991). They are designed by molecular
modeling to provide the proper spacing between portions of an
oligonucleotide which are to interact with different strands of a
double-stranded DNA molecule. Examples of oligonucleotides having
such linkers include the following: TTGCCTTGCCT-switchback
linker-CCT-switchback linker-CTTGCCT (corresponding to nucleotides
105-125 of the double-stranded sequence represented by SEQ ID NO:1)
or portions thereof.
Single-stranded, linear or circular oligonucleotides containing
nucleotide analogs which are able to complex specifically with all or
part of a p53-specific binding site as described above are also
contemplated as part of the invention. (See Kool, J. Am. Chem. Soc.,
vol. 113:625-626, 1991, with regard to circular oligonucleotides.) Such
oligonucleotides containing nucleotide analogs should comprise at least
about ten nucleotides in length in order to have the requisite
specificity with respect to the entire human genome. Oligonucleotides
or oligonucleotides containing nucleotide analogs which are able to
complex specifically with part of a p53 binding site as well as with
adjacent sequences are also contemplated as part of the invention. The
oligonucleotides or oligonucleotides containing nucleotide analogs will
preferably bind to the identified p53 binding regions. However, other
binding regions in the human genome may well be found which are also
suitable targets for the oligonucleotides or analogs of the present
invention. These other binding sites may well be the primary targets of
p53; complexation of these sites may inhibit the unregulated growth
which characterizes neoplastic cells.
Double-stranded DNA fragments which comprise a p53-specific
DNA binding site and are attached to an insoluble polymeric support
are also contemplated by this invention. The support may be agarose,
cellulose,. polycarbonate, polystyrene and the like. Such supported
fragments may be used in screens to identify compounds which bind to



2070979
p53-specific DNA binding sites. Similarly, such supported fragments
may be used to perform diagnostic tests on cell lysates from suspected
tumor tissues. They may also be used in assays used to screen potential
chemotherapeutic agents, as discussed infra.
Although any method can be employed which utilizes the
p53-specific DNA binding sites of the present invention, two particular
methods are disclosed for screening for additional compounds that bind
to p53-specific DNA binding sites. According to one method a test
compound is incubated with a supported DNA fragment, as described
above. The amount of test compound which binds to the supported
DNA fragment is determined. This determination can be performed
according to any means which is convenient. For example, the amount
of a compound which can be removed of ter incubation with the
supported fragment can be ~ compared to the amount originally applied.
Alternatively, the test compound can be labelled and the amount which
binds to the supported fragment can be assayed directly. In order to
render this screening method more specific, soluble DNA fragments
which do not contain the p53 DNA binding sequence can be added to
the incubation mixture. The soluble fragments would not have the
ability to specifically bind to p53 wild-type protein.
According to another screening method for compounds to
simulate the specific DNA binding activity of p53, test compounds are
incubated with supported DNA fragments as described above.
However, in this method wild-type p53 protein is also added to the
incubation mixture. The amount of p53 protein which binds to the
DNA fragment is measured using methods as described above. The
amount of p53 protein bound is compared to the amount which binds in
the absence of the test compound. Any diminution of p53 binding
which results from the presence of the test compound is presumptively
due to the competition of the test compound with p53 for the specific
DNA binding sites of the supported fragments. Direct binding of the
test compound to the binding site fragments can be confirmed using the
assay described above.
The presence or absence of p53 genes or gene products can also
be detected in body samples, such as, serum, stool, or other body fluids,



2070979
-19-
such a~ urine and sputum. The same techniques discussed above for
detection of the presence or absence of gene products in tissues can be
applied to other body samples. By screening such body samples, a
simple early diagnosis can be achieved for many types of cancers. In
addition, the progress of chemotherapy or radiotherapy can be
monitored more easily by testing such body samples for the presence or
absence p53 genes or gene products.
The methods of the present invention for diagnosis and therapy
of neoplastic tissue is applicable across a broad range of tumors. These
include lung, breast, brain, colorectal, bladder, mesenchyme, prostate,
liver as well as stomach tumors. In addition the method may be used in
leukemias and osteosarcomas. It thus appears that the p53 gene has a
role in the development of a broad range of tumors. The methods of
diagnosis and therapy of the present invention are applicable to any
tumor in which p53 has a role in tumorigenesis. The diagnostic method
of the present invention is useful for clinicians so that they can decide
upon an appropriate course of treatment. For example, a tumor
displaying loss of both p53 alleles suggests a more aggressive
therapeutic regimen than a tumor displaying loss of only one p53 allele.
Compounds which have p53-specific DNA-binding activity,
including wild-type p53 protein, polypeptides corresponding to portions
of wild-type p53 protein, oligonucleotides and oligonucleotide
containing nucleotide analogues, as well as other organic molecules can
also be administered to humans and animals as a pharmaceutical and
therapeutic composition. Effective amounts will be administered to
cause neoplastic cells to become less aggressively neoplastic or even to
stop the growth of the neoplastic cells entirely. Generally, such
amounts will be in the range of 10 ng to 10 ug per dose per person or
other animal. The therapeutic compounds can be prepared in any
conventional pharmaceutical excipient, such as physiological saline or
other physiologically compatible aqueous buffer. Typically, the
compounds will be administered by injection, either intravenous or
intramuscular. However, other administration methods as are known in
the art and may be used to administer the compounds of the present
invention.




v ~. 24'~4~~9
-20-
As a result of the discoveries of the present invention, screening
methods can be devised to isolate chemical agents which may have use
in cancer therapy. Specifically, agents can be screened for the ability
to affect the structure of mutant p53 molecules so that their ability to
bind and/or transactivate at specific-DNA-binding sites is restored.
The necessary components for s~~ch a screening method are provided by
this invention and include DNA molecules which contain more than one
monomer of the sequence RRRCWWGYYY, and mutant p53 proteins
which are found in tumors.l/
One such prescreening method is a binding assay in which the
amount of binding of a p53 mutant protein to a DNA molecule which
comprises the consensus binding site (or a conforming sequence) is
measured. The amount of binding is also measured for a p53 mutant
protein in the presence of a test substance. If the test substance
increases the amount of p53 binding, then the test substance is a
candidate for use in anti-tumor therapy. Further testing will be
desirable before use in humans is attempted.
Methods for measuring the amount of binding can be any which
are known in the art. See, e.g., Tan, et al., Cell, 62:367-3Z? (1990).
One particular method employs immunoprecipitation. Briefly, purified
p53 or a lysate of a cell expressing p53 is incubated with radiolabeled
DNA and anti-p53 antibodies under conditions where proteins bind to
DNA. Protein A-Sepharose and poly-dIdC-poly-dIdC are then added for
an additional incubation. A pellet is formed and washed and the
proteins are removed by digestion with a protease, and DNA is obtained
by phenol extraction. The extracted DNA is then analyzed by
electrophoresis and quantified. Quantitation of the DNA can be by
autoradiography, for example. The amount of DNA
immunoprecipitated is proportional to the amount of binding of the p53
protein to the DNA.
1/ Not all mutations in p53 destroy specific-DNA-binding ability.
For example, mutations in phosphorylation sites of p53 have
been made and tested; they retain binding activity. Such
mutations have never been found in tumors. Mutations in p53
which are found in tumors are termed oncogenic herein.



2070979
-21-
According to another method, the ability of a mutant p53
protein from a tumor to transactivate transcription in vitro is assessed
with and without a test substance. If the test substance increases the
amount of transcription activated by the p53, then the test substance is
a candidate for use in anti-tumor therapy. Transcriptional activation is
measured using a transcription construct which comprises a reporter
gene encoding a convenient assayable enzyme activity, such as
chloramphenicol acetyltransferase or S-galactosidase, and an upstream
p53 consensus binding site (or a conforming sequence). The binding site
must be upstream, although the distance from the start of transcription
is not critical. The binding site, which is adjacent to the reporter gene,
may be from 0 to 1 kb upstream. In vitro transcription assay systems
are well known in the art. See, e.g., Lue, Science, 246, 661-664 ( 1989).
According to still another method, transcriptional activation can
be measured in a cell containing a mutant p53 protein which has been
transfected with a reporter gene construct comprising a gene encoding
an assayable product, such as an enzyme or antigen, with a p53
consensus binding site (or conforming sequence) adjacent and upstream
therefrom. The transfected cells are treated with a test substance. If
the amount of transactivation caused by the mutant p53 is enhanced by
the test substance, then the substance is a candidate for anti-tumor
therapy presumably due to its interaction with the mutant protein. A
particular transactivation assay (transient expression assay) is
described in Example 12, inf ra. Particular reporter constructs are
shown in Figure 12A. Others may be used within the scope of the
invention. Alternatively, if the transfected cell contains wild-type p53
diminished activation would indicate that the test substance competes
with p53 for binding sites.
In another embodiment of the invention, oligonucleotides can be
isolated which restore to mutant p53 proteins the ability to bind to the
consensus binding sequence or conforming sequences. Mutant p53
protein and random oligonucleotides are added to a solid support on
which p53-specific-binding DNA fragments are immobilized.
Oligonucleotides which bind to the solid support are recovered and
analyzed. Those whose binding to the solid support is dependent on the

CA 02070979 2003-07-03
-22-
presence of the mutant p53 protein are presumptively binding the support by
binding
to and restoring the conformation of the mutant protein.
Transient expression constructs are conveniently made on, plasmids and viral
vectors, so that they can be propagated. These can also be used in vitro for
transcription assays in the presence of RNA polymerase, ribonucleotides, and
other
cofactors.
EXAMPLES
Example 1
This example demonstrates the screening methods used to identify a p53
sequence-specific binding site.
In an attempt to identify a possible sequence-specific binding site, numerous
cloned DNA sequences were screened using an immunoprecipitation technique. The
immunoprecipitation assay for, DNA-binding was modified from R.D.G. McKay, J.
Mol. Biol. 145. 471 (1981). Binding reactions included S ~1 of vaccinia-
infected cell
lysate or purified baculovirus p53 preparations, 95 ~l binding buffer (20 mM
Tris pH
7.2, 100 mM NaCI, 10% glycerol, 1 % NPT""-40, S mM EDTA), 6-20 x 104 dpm 3zp-
labelled DNA, 4 ~1 (0.4 pg) pAb421 and 4~1 (0.4 ug) pAb18O1 anti-p53 purified
monoclonal antibodies (Oncogene Science), at 4°C for 30 min. 1.5 mg
protein A-
SepharoseT"~ (Sigma) and 12.5 ~g poly-dIdC-poly-dIdC (Pharmacia), in 25 ~1
binding
buffer, were added and the reactions rotated end-over-end at 4° C for
30 min. The
pellet was washed twice with binding buffer and the proteins digested with SDS-

proteinase K, then extracted with phenol and chloroform. The DNA was ethanol-
precipitated and dissolved in electrophoresis sample buffer. The fragments
were
separated on a Tris-borate nondenaturing polyacrylamide (7, 10, or 12%) gel.
The gel
was fixed and dried for autoradiography.
Recombinant vaccinia viruses expressing p53 from the 40K promoter were
isolated and purified as described in J. Lyons et al., Infec. and Immun. 58
4089
( 1990).
Vaccinia-infected cell lysates were prepared as follows: the indicated cells
(about 2.5 x 10') were infected (MOI 2), harvested at 24 hrs and lysed in 2 ml
lysis
buffer (PBS containing SmM EDT'A, 0.5%




2070979
-23-
NP-40, . 0.5 mM PMSF, 10 ug/ml TPCK, 1 ug/ml aprotinin, 10 ug/ml
trypsin inhibitor, 1 ug/ml leupeptin). Cell debris was pelleted at
16,OOOxg and the supernatant UV-irradiated (0.5 joules). The
supernatant was frozen in aliquots at -80°C and found to be stable for
at least 6 months.
Baculovirus-produced p53 was purified as described in P.N.
Friedman, et al., Proc. Natl. Acad. Sci. USA, 87, 9275 (1990); while the
presence of an accessory cofactor in the purified preparation cannot be
excluded, SDS-polyacrylamide gel electrophoresis of the preparation
revealed only a single polypeptide migrating at 53 kD upon
silver-staining.
Two classes of clones were tested. The first class consisted of
400 clones containing inserts of 300 to 1000 base-pairs obtained
randomly from the human genome. The second class consisted of
cosmid and plasmid clones chosen because they contained sequences
which might be important in normal growth control. Random human
genomie clones were prepared from a partial MboI digest of human
DNA; 300-1000 by fragments were purified and ligated to EcoRI linkers,
then cloned into the EcoRI site of pBluescript II (Stratagene). Clones
selected because they might be important in normal growth included
SP65hFosAva2, containing regulatory sequences from the fos gene,
obtained from T Curran; 772 CBE (J.E. Sylvester, R. Petersen, R.D.
Schmeckel, Gene, 84, 193 (1989)), from J. Sylvester; a 4.4 kb BgIII
subclone of Lambda 5R (R. Misra, et al., Nucl. Acids Res. 17:8327
(1989)), from C. Schmid; the c-myc clone was HSR-1 (K. Alitano, et al.,
Proc. Natl. Acad. Sci. USA, 80, 1707 (1983)) from J. Bishop. Cosmids
containing the entire p53 genomic region were from Y. Nakamura;
cosmids including the entire DHFR amplicon from hamster cells (B.
Anachkova and J.L. Hamlin, Mol. Cell Biol. 9, 532 (1989)) were from J.
Hamlin.
Each clone was digested with an appropriate restriction
endonuclease, end-labelled with 32P, and incubated with p53 protein
from a lysate of cells infected with a recombinant vaccinia virus
expressing p53 protein. Labelled DNA fragments which bound to p53
were then recovered by immunoprecipitation with monoclonal




20709?9
-24-
antibodies against p53. Of the more than 1400 restriction fragments
tested, only two bound reproducibly to p53 under the experimental
conditions used: a 259 basepair HinfI fragment (fragment A) of clone
7'12 CBE (Panel 2, Fig. lA), and a 190 basepair HinfI fragment (fragment
B) of clone Lambda 5R (Panel 3, Fig. lA); these fragments bound to a
far greater extent than any of the other labelled fragments of larger or
smaller size present in the same assay mixes.
Example 2
This example demonstrates that the immunoprecipitation of
fragment A is dependent on both p53 protein and anti-p53 antibodies.
The immunoprecipitation assay was performed on fragment A as
described in Example 1. Cell lysates were produced by infection with
vaccinia virus that did or did not contain an insert of wild-type p53
DNA. Either anti-p53 antibodies were used or normal mouse IgG was
used.
Lysates from cells infected with wild-type vaccinia virus (devoid
of p53) were not able to specifically immunoprecipitate fragment A
(Fig. iB). Similarly, the detection of the precipitation of fragment A
was dependent on the presence of anti-p53 antibodies (Fig. iB). The
binding was evident in lysates prepared from either human HeLa cells
or monkey BSC40 cells infected with vaccinia virus and expressing
wild-type p53 (Fig. iB).
Affinity-purified baculovirus-produced wild-type p53 protein
was substituted for the vaccinia-infected cell lysates in the
immunoprecipitation assay and found to bind fragment A strongly (Fig.
2A). This suggested that the binding to fragment A was an intrinsic
property of the p53 polypeptide and not dependent on other factors
present in the vaccinia virus-infected cell lysates.
Example 3
The example demonstrates that p53 mutant proteins found in
human tumors fail to bind to fragment A.
Increasing quantities of wild-type and mutant 273hig p53
protein, affinity purified from a baculovirus expression system, were
used to immunoprecipitate labelled fragments from CBE. See Fig. 2A.
The proportion of fragment A bound to wild-type p53 protein increased



2070979
-25-
in tandem with the amount of p53 added to the assay mixture. (Fig.
2A) In contrast, fragment A did not specifically bind to a mutant form
of p53 (273hig) protein even at the highest p53 protein concentration
used. The 2?3hig mutation is the most common p53 mutant identified
in human tumors. Another p53 mutant (1?Ship) protein commonly
found in human tumors also failed to bind to fragment A (Fig. iB).
Example 4
This example defines the particular sequences within fragment
A that allow it to bind to wild-type p53 protein.
Fragment A was subcloned, and the 259 by insert from the
subelone (lOd, SEQ ID: 1 and Fig. 3A) bound to p53 as expected. A
strategy based on the polymerise chain reaction (PCR) and restriction
endonuclease digestion was used to generate subfragments of this
clone.
One primer for each PCR was labelled with 32P at the 5' end
with T4 polynucleotide kinase in a 5 ul reaction, and the kinase
inactivated at 70 ° C for 5 min. PCR contained 350 ng of each of the
appropriate primers and approximately 50 ng plasmid template in a 50
ul reaction, using 25 cycles and the PCR conditions specified in Baker
SJ, et al., Cancer. Res., 50:7717 (1990). The products were extracted
with phenol and chloroform, ethanol-precipitated, and dissolved in
3mM Tris, 0.2 mM EDTA prior to binding. Subfragment 1 contained by
1 to 425 of subclone lOd of fragment A (Fig. 3A); subfragments la, ib,
ic, id, and le were generated by digestion of subfragment 1 with
BamHI, MboI, HindIII, HindIII, and BamHI, respectively, from fragment
1. Subfragment 2, contained by 283 to 425. Subfragment 3a was
generated by digestion of subfragment 3 (bp 106 to 294) with Hae III.
Subfragment 4a was produced from subfragment 4 (gp 1 to 141) by Hind
III digestion. Subfragments 5a and 5b were products of the HaeIII
digestion of subfragment 5 (bp 87 to 141). "Mutant" subfragments
Smutl and 5 mutt were produced using primers P3m1
(5'-GAAAGAAAAGGCAAGGCCAGGAAAGT-3') and P3mut2
(5'-GAAAGAAAAGGCAAGGCCATTAAAGT-3') and were identical to
subfragment 5 except for the positions underlined in the primers.
Subfragment 6 contained by 106 to 138, and the insert was excised by




2070979
-26-
restriction with HindIII and BamHI to generate 6a or with HindIII and
EcoRI to generate 6b. Subfragment 3, including basepairs 106 to 294
(Fig. 4B, panel 2) bound well to p53 as did subfragment 4, containing
basepairs 1 to 141 (Fig. 4B, panel 3). This and similar assays done with
additional subfragments (Figs. 4A and 4B) localized the critical
sequences to basepairs 106 to 141. This segment contained three
repeats of the sequence TGCCT (Fig. 3A). Digestion of subfragment 3
with HaeIII (cleaving between by 125-126 and removing two of the
TGCCT repeats) greatly reduced this binding (Fig. 4B, subfragment 3A,
panel 2), suggesting that a critical sequence lay at or near this
restriction site and that a single TGCCT repeat was not sufficient for
binding. Additional subfragments were tested (#5, by 87 to 141, Figs.
4A and 5B; #6, by 106 to 138, Figs. 4A and 4B, panel 4), and it was
established that a 33 by insert containing three TGCCT repeats
provided binding capability.
Example 5
This example demonstrates that certain G residues are critical
for binding of p53 to fragment A.
We studied the requirements for binding at the single nucleotide
level using a methylation interference assay. PCR products labelled at
one end were generated using primers labelled with T4 polynucleotide
kinase (US Biochemicals). The product of each PCR reaction was
purified by polyacrylamide gel electrophoresis, eluted from a crushed
gel slice in 500 mM ammonium acetate, extracted with phenol and
chloroform, and precipitated with ethanol. 2 x 106 dpm of DNA was
methylated at guanine residues using dimethylsulfate as described (T.
Maniatis, E.F. Fritsch, J. Sambrook, Molecular Cloning A Laboratory
Manual, 1st ed., Cold Spring Harbor Laboratory, 1983, p. 4'17),
ethanol-precipitated and dissolved in 10 ul 3 mM Tris, 0.2 mM EDTA.
0.5 ul was removed as the DNA control. 4.5 ul was added to a binding
reaction containing baculovirus-produced p53 or vaccinia-infected cell
lysates. The precipitated DNA was purified by SDS/proteinase K
digestion, extracted with phenol-chloroform, and ethanol-precipitated.
The control DNA and precipitates of bound DNA were cleaved with
piperidine at the methylated sites. Equivalent amounts of labelled




2070979
-2?-
DNA were loaded and separated on a denaturing polyacrylamide (14.596)
sequencing gel, which was fixed and dried for autoradiography.
Subfragment 5, demonstrating efficient binding (Figs. 4A and SB) was
methylated in vitro, and immunoprecipitated of ter binding to p53. The
bound DNA was then cleaved with piperidine at methylated residues
and separated by electrophoresis on a sequencing gel. Assay of one
strand (Fig. 5A, right) demonstrated that methylation at the G at by
120 significantly interfered with binding. On the opposite strand (Fig.
5A, left), the most effective interference was produced by methylation
at G residues at by 11'1, 121, and 122. Partial interference was also
produced by methylation at nearby G sites (bp 110-112, 114, and 115).
Thus, the methylation interference assay pinpointed one of the central
TGCCT repeats (centered at basepair 121) and adjacent basepairs as
critical for binding.
To obtain independent evidence of the specificity for the G
residues identified by methylation interference, in vitro mutagenesis
was used. A DNA fragment was generated that was identical to
subfragment 5 except for the substitution of G at by 120, 121, and 122
with T residues. This ~tmutant~~ subfragment (#5mut2) bound poorly to
p53 (Fig. 5B). A fragment identical to subfragment 5 except for a
single basepair (T substituted for G at by 120) was then tested. This
fragment (#5mut1) also did not bind appreciably (Fig. 5B).
Example 6
This example defines the region of fragment B which is
important for p53 binding.
Fragment B was subcloned (sequence in Fig. 3B and SEQ ID
N0:2). Interestingly, this fragment had two repeats of the TGCCT
motif (centered at by 135 and 152). A PCR strategy similar to that
used for fragment A was used to demonstrate that a 95 basepair
subfragment (bp 104 to 198), which contained both of these repeats,
was sufficient for binding (Fig. 4B, panel 5).
Example Z
This example shows that expression of the wild-type p53 gene in
human colorectal carcinoma cells dramatically inhibits their growth




2070979
-28-
and that a mutant p53 gene cloned f rom a human colorectal carcinoma
was incapable of exerting such inhibition.
The colorectal carcinoma lines SW480 and SW83?, which are
representative of 7596 of colon carcinomas, have each lost one copy of
chromosome 1?p (including the p53 gene) and the remaining p53 allele
is mutated (Baker, et al., Science 244, 217 (1989); Nigro et al, Nature
342, 705 (1989)). The SW837 line contains an arginine to tryptophan
mutation at codon 248 (Nigro, supra). The SW480 line contains two
point mutations, arginine to histidine at codon 273 and proline to serine
at codon 309 (Nigro, supra.). The substitutions at codon 248 and 273 are
typical of those observed in human tumors, occurring within two of the
four mutation "hot spots" (Nigro, supra).
For the transfection studies, we constructed a vector,
pCMV-Neo-Bam, engineered to contain two independent transcription
units. The expression vector pCMV-Neo-Bam was derived from plasmid
BCMGNeo-mIL2 (Karasuyama, et al., J. Exp. Med. 169, 13 (1989) by
excision of the human beta globin sequences and bovine papilloma virus
sequences with Bam HI and Not I. Next, the interleukin 2 (IL-2)
sequences present at the unique Xho I site were removed, and the Xho I
site was changed to a Bam HI site by linker addition. The vector
included CMV promoter/enhancer sequences, which could drive
expression of the insert at the Bam HI site, and splicing and
polyadenylation sites derived from the rabbit beta globin gene, which
ensured proper processing of the transcribed insert in the cells. A
pBR322 origin of replication and s-lactamase gene facilitated growth
of the plasmid in E. coli. The plasmid conferred geneticin resistance
through expression of the neomycin resistance gene under separate
control of an HSV thymidine kinase promoter. The first transcription
unit comprised a cytomegalovirus (CMV) promoter/enhancer upstream
of a site for insertion of the cDNA sequences to be expressed, and
splice and polyadenylation sites to ensure appropriate processing. The
second transcription unit included a herpes simplex virus (HSV)
thymidine kinase promoter/enhancer upstream of the neomycin
resistance gene, allowing for selection of transfected cells in geneticin.




2070979
-29-
wild-type p53 cDNA was inserted into pCMV-Neo-Bam to
produce pC53-SN3. Similarly, a vector, pC53-SCX3, expressing a
mutant eDNA from human colorectal tumor CX3, was also constructed.
The only difference between pC53-SN3, and pC53-SCX3 was a single
nucleotide (C to T) resulting in a substitution of alanine for valine at
p53 codon 143 in pC53-SCX3. The two constructs were made as
follows: a 1.8-kb Xba I fragment, extending from nucleotide -130 to
1671 relative to the translation initiation site, was isolated from
wild-type or CX3 cDNA clones. The fragment was blunt-ended with
the Klenow fragment of DNA polymerase, ligated to Bam HI linkers,
and cloned into the unique Bam HI site in the expression vector
pCMV-Neo-Bam.
The constructs were transfected into SW83? and SW480 cells
(obtained from the American Type Culture Collection, Rockville,
Maryland), and geneticin-resistant colonies were counted 3 weeks later.
Cells transfected with pC53-SN3 formed five-to tenfold fewer colonies
than those transfected with pC53-SCX3 in both recipient cell types
(Table 1).
Table 1. Colony formation after transfection with wild-type
and mutant p53 expression vectors. For each experiment, one or
two 75-cm2 flasks were transfected, and the total colonies
counted after 3 to 4 weeks of selection in geneticin (0.8
mg/ml). Exp. experiment.
No. of geneticin-resistant


colonies formed


Cell line Exp. ______________________________


pC53-SCX3 pC53-SN3


(mutant) (wild-type)


SW837 1 754 66


2 817 62


SW480 1 449 79


2 364 26


RKO 1 1858 190


2 1825 166


VACO 235 1 18 16


2 26 28





2070979
-30-
In both.SW83? and SW480 cells, the number of colonies produced by the
expression vector pCMV-Neo-Bam (without a p53 eDNA insert) was
similar to that induced by the pC53-SCX3 construct.
These results suggest that the wild-type p53 gene inhibits the
clonal growth of both the SW83? and SW480 cell lines; however, a
significant number of colonies formed after transfection of the
wild-type construct. If wild-type p53 expression were truly inhibitory
to cell growth, one would expect that no colonies would form or that
p53 expression in the colonies that did form would be reduced
compared to that produced with the mutant p53 eDNA construct. To
evaluate this issue, we expanded independent SW480 and SW83?
colonies into lines, and ribonuclease (RNase) protection analysis was
performed to determine the amount of p53 mRNA expressed from the
exogenously introduced sequences. Twelve of 31 lines (3896) derived
from transfection with the pC53-SCX3 construct were found to express
the exogenous mutant p53 mRNA. This percentage was consistent with
results expected in human cells transfected with a vector containing
two independent transcription units. Previous studies have shown that,
in contrast to rodent cells, primate cells are able to integrate only a
small amount of foreign DNA (approximately 6 kb), so that only 10 to
3096 of clones selected for the expression of one transcription unit also
contain the second unit in an intact form (F. Collabere-Garapin, et al.,
Gene 50, 2?9 (1986); Hoeijmakers, et al., Exp. Cell Res., 169, 111 (1987);
Mayne et al., Gene 66, 65 (1988), Dean, et al., Exp. Cell Res., 183, 4?3
(1989). In contrast, no expression of exogenous p53 wild-type mRNA
was seen in any of 21 clonal lines established from either SW480 or
SW83? cells transfected with the pC53-SN3 vector (Fig. 6A). These
RNase protection results were supported by analysis of the exogenous
p53 DNA sequences within the clones. All of the p53-expressing clones
derived from the pC53-SCX3 transfection contained an intact copy of
the exogenous p53 gene (Fig. 6B). In contrast, in all the clones derived
from the pC53-SN3 transfection, the exogenous p53 sequences were
deleted or rearranged (Fig. 6B).
The results from individual clones were further supported by the
analysis of pooled clones, in which numerous colonies could be



2070979
-31-
simultaneously assessed. Forty or more clones f rom two to three
separate transfection experiments were pooled and analyzed
approximately 3 weeks after transfection. RNase protection studies
showed substantial expression of wild-type sequences was not
detectable (Fig. TA). Results from Southern (DNA) blotting were
consistent with the RNase protection studies, in that pooled colonies
from the wild-type transfectants had no detectable unrearranged
exogenous p53 sequences, in contrast to the intact p53 sequences in
colonies derived from the mutant p53 cDNA expression vector (Fig.
?B).
The conclusions made from the above experiments are
dependent on the assumption that p53 protein was produced in the
transfeeted cell lines. Clones containing exogenous mutant p53
sequences produced p53 mRNA at a concentration 1.5 to 3.5 times
higher than that produced by the endogenous p53 gene (Figs. 6A and
7A). Immunoblot analysis showed that there was a concomitant small
increase in p53 protein expression in the transfectants (1.5- to 3-fold)
compared to the untransfected cells. However, this increase was
difficult to measure quantitatively, since these cells produced
significant amounts of endogenous p53 protein that (unlike endogenous
p53 mRNA) could not be distinguished from that produced by the
vectors. To confirm that transfeeted human cells expressed p53
protein from our constructs, we studied an additional colorectal
carcinoma cell line (RKO). RKO cells were obtained through the
generosity of M. Brattain. Although RKO cells did not contain a
mutation within the susceptible p53 coding sequences, i.e., exons 5-9,
they expressed low concentrations of p53 mRNA compared to normal
colorectal mucosa or the other lines studied and did not produce
detectable amounts of protein.
Results of colony formation assays in transfected RKO cells
were similar to those in SW480 and SW837 cells. Colony formation by
wild-type p53 gene transfectants occurred with a tenfold decrease in
efficiency compared to the mutant p53 construct (Table 1).
Immunocytochemical detection of p53 protein in transfected RKO cells
was done as follows: approximately 5 x 10° cells were cytocentrifuged


CA 02070979 2003-07-03
-32-
onto polylysine-coated slides, fixed for 10 min in formalin, and permeabilized
for 5
min in 0.5% TritonT"" X-100. A mouse monoclonal antibody against human p53
protein (Ab 1801 ) in combination with the ABC immunoperoxidase system (Vector
Laboratories), was used for immunocytochemical detection of p53 protein
(Banks, et
al., Eur. J. Biochem. 159, 529 (1986)). Ten to 20 randomly selected
microscopic
fields were analyzed per slide. These observations are consistent with the
greater
stability of mutant compared to wild-type p53 protein noted previously (C.A.
Finlay
et al., Mol. Cell Biol. 8, 531 (1988)). However, transient mRNA expression was
also
significantly lower in the SN3 transfectants compared to the SCX3
transfectants at 48
and 96 hours, supporting the idea that RKO cells expressing wild-type p53 were
at a
selective disadvantage compared to those producing mutant p53 products.
To obtain additional evidence that cells expressing p53 were inhibited in
their
growth potential, we examined the effect of p53 gene expression on DNA
synthesis in
transfected RKO cells were labeled with [3H] thymidine for 2 hours. The cells
were
subsequently fixed, immunocytochemically stained for the presence of p53
protein,
and autoradiographed. The number of cells undergoing DNA replication was only
slightly lower in cells producing exogenous mutant p53 protein than in cells
that did
not express any detectable p53 protein. Expression of the wild-type protein,
however,
dramatically inhibited the incorporation of thyrnidine (Table 2).
Table 2. Immunocytochemistry and [3H]thymidine incorporation of transfected
RKO
cells.
Percent of cells Percent of cells


expressing p53 incorporating


protein at [3H]thymidine in


Plasmid _______________________________________
___________________________________


6 24 48 96 p53 p53 non-


hrs hrs hrs hrs expressors expressors


pC53-SCX3 1.0 11 4.3 2.0 24 31


pC53-SN3 1.9 5.2 0.3 0.2 1.7 33






2070979
-33-
These results all suggested that wild-type p53 exerted an
inhibitory effect on the growth of carcinoma cells in vitro. To
evaluate whether this inhibitory effect was cell type-specific, we
transfected colorectal epithelial cells derived from a benign tumor of
the colon (the VACO 235 adenoma cell line). VACO 235 cells are
described by J.K.V. Willson et al., Cancer Res., 47, 2704 (1987).
Previous studies have shown that most adenomas contain two copies of
chromosome 1?p and express wild-type p53 mRNA at concentrations
similar to that of normal colonie mucosa. Analogously, the p53 alleles
of the VACO 235 cell line were sequenced (exons 5-9) and found to be
wild-type and the expression of p53 mRNA was found to be similar to
that of normal colorectal mucosa. In contrast to the results seen with
SW480, SW83?, and RKO cells, the pC53-SN3 and pC53-SCX3
constructs produced similar numbers of geneticin-resistant colonies
after transfection of the VACO 235 line (Table 1). We considered,
however, that the most definitive test for differential growth
inhibition by wild-type versus mutant p53 genes involved analysis of
exogenous p53 expression in pooled transfectants. Through such
analysis, a large number of colonies could be examined simultaneously
and the expression of exogenous mutant and wild-type p53 genes
directly compared. Striking differences in the relative expression from
the transfected genes were seen in all three carcinoma cell lines
tested. VACO 235 transfectants, however, expressed similar amounts
of exogenous p53 mRNA from either pC53-SN3 (wild-type) or
pC53-SCX3 (mutant) p53 constructs (Fig. 7A).
In summary, our results suggest that expression of the wild-type
p53 gene in colorectal carcinoma cell lines was incompatible with
proliferation. The inhibitory effects of wild-type p53 were specific in
two ways. First, a single point mutation in a p53 gene construct
abrogated its suppressive properties as measured by three separate
assays (colony formation, exogenous p53 expression in transfected
clones, and thymidine incorporation). The CX3 mutant provided a
control for gene specificity as it contained only one conservative
mutation, resulting in a substitution of one hydrophobic amino acid
(alanine) for another (valine) at a single codon. Second, the




. ~ 2070979
-34-
growth-suppressive effect of the wild-type p53 construct was cell
type-specific. Introduction of the wild-type vector into the VACO 235
adenoma cell line had no measurable inhibitory effect compared to the
mutant p53 vector. There are several differences between the cell
lines that could account for the differential effect of the introduced
vectors. Regardless of the basis for the difference, the results with the
VACO 235 cell line minimize the possibility that the wild-type p53
construct had some nonspecific, toxic effect on recipient cells; the
effect was cell type-dependent.
The transfection and expression results of Table 1 and Fig. 7A
suggest that cells at the premalignant stages of tumor progression
(VACO 235) may be less sensitive to the inhibitory effects of wild-type
p53 than malignant cells (SW480, SW83?, and RKO). This hypothesis is
consistent with previous results that suggest the wild-type p53 is less
inhibitory to the growth of normal rat embryo fibroblasts than to their
oncogene-transfected derivatives. Finlay et al., Cell 57, 1083 (1989);
Eliyahu et al., Proc. Natl. Acad. Sci. USA, 86, 8763 (1989). This
sensitivity may only be relative: expression of the wild-type gene at
high concentrations might inhibit the growth of any cell type, including
non-neoplastic cells, by overwhelming normal regulatory processes
such as phosphorylation. Samad et al., Proc. Natl. Acad. Sci. USA, 83,
897 (1986); Meek et al., Mol. Cell Biol. 8, 461 (1988). Genetic
alterations that occur during the progression of colorectal tumors may
increase the sensitivity of cells to p53 inhibition, making wild-type p53
expression a key, rate-limiting factor for further tumor growth and
expansion. At this point, and not before, mutations in the p53 gene
would confer a selective growth advantage to cells in vivo, which
would explain the frequent occurrence of p53 gene mutations and
allelic loss only in the more advanced stages of colorectal
tumorigenesis.
Example 8
This example demonstrates the identification of human genomic
fragments that can bond to wt p53 protein in vitro.
This schema in Figure 8A outlines the experimental strategy
used for the isolation and analysis of p53 binding sites. Total human


CA 02070979 2003-07-03
-35-
genomic DNA fragments were ligated to specially designed "catch" linkers
(Kinzler,
et al. (1989), Nucleic Acids Research, 17:3645-3653, and Kinzler, et al.
(1990),
Molec. Cell. Biol., 10:634-642) to allow for subsequent PCR amplification and
cloning. The linked genomic DNA was then incubated with wt p53 and
precipitated
with anti-p53 antibodies. The bound DNA was subsequently amplified by PCR
using
primers complementary to the catch linkers, and the process repeated. After
four
rounds of sequential immunoprecipitation and PCR, the amplified and selected
(AS)
DNA was cloned. Clones were picked at random and tested for p53 binding first
by
immunoprecipitation (IP), and then by methylation interference (MI) and DNase
I
protection (DP).
Following the outline in Figure 8A., we tested the inserts of 530 clones for
binding to p53. Restriction fragments of the clones were end-labeled and
incubated
with purified human wt p53 protein produced in baculovirus-infected cells.
Whole-genome PCR was performed as previously described, except that only
one oligonucleotide (5'-GAGTAGAATTCTAATATCTC-3') was used for
amplification (Kinzler, et at. (1989), Nucleic Acids Research, 17:3645-3653,
and
Kirizler, et al. (1990), Molec. Cell. Biol., 10:634-642). Two hundred ng of
"catch"-
linked human genomic DNA were incubated with 100 ng of baculovirus-produced
human wt p53 purified as described (Friedman, et al (1990), Proc. Natl. Acad.
Sci.
U.S.A., 87:9275-9279), and immunoprecipitated as described below. After 4
rounds
of IP and PCR, the AS DNA was cleaved with Eco RI and cloned into either the
vector Lambda ZapT"" II or pBluescriptT"" II SK+ (Stratagene). Individual
clones were
picked at random and tested for p53 binding. In panel B, cloned plasmid DNA
samples were cleaved with Eco RI and end-labeled by Klenow fill-in. For IP
(McKay, et al. (1981), J. Mol. Biol, 145:471-479), ten ng of DNA were
incubated
with 100 ng of baculovirus-produced human wt p53 and 100 ng of poly dI-dC at
4°C
for 30 minutes in 100 pl of "DNA-binding buffer" containing 100 mM NaCI, 20 mM
Tris, pH 7.0, 10% glycerol, 1 % NP40, and 5 mM DTT. DNA fragments bound to
p53 were complexed to antibodies by the addition of 8 p.l containing 400 ng
each of
anti-p53 antibodies pAb421 and pAb1801, both obtained from




2070979
-36-
Oneogene Science, and incubated for 30 minutes at 4 ° C. The
DNA-binding buffer containing 1.5 mg protein A precipitated following
the addition of 26 ul of DNA-binding buffer containing 1.5 mg protein A
Sepharose and 10 ug of poly dI-dC and mixing at 4 ° C for 30
minutes.
After removal of the supernatant, the immunoprecipitate was washed
twice with 1 ml of DNA-binding buffer. Bound DNA was purified by
treatment with SDS and proteinase K at 48 ° C for 30 minutes,
extracted with phenol and chloroform, precipitated with ethanol,
separated by electrophoresis on a 1096 nondenaturing polyacrylamide
gel, and autoradiographed.
Twenty-three of the clones were found to contain fragments
that bound to p53. Examples of the IP experiments are shown in Figure
8B. Clone S61 (lanes iIB,C) contains a single genomic fragment of 202
by which bound to p53. Clone N2 contained five fragments, only one of
which (357bp) bound to p53 (lanes lOB,C). Other examples of
p53-binding fragments were obtained, and each of these was subcloned
for further analysis. In contrast, we found that none of over 1000
clones containing unselected human DNA inserts of similar size bound
to p53 using the IP assay. Thus, the whole-genome PCR procedure
significantly enriched for p53-binding sequences. -
Example 9
This example demonstrates the localization of p53 contacts with
bound DNA fragments.
Localization of the regions bound by p53 was obtained by DP or
MI assays using the subcloned DNA fragments as probes. For MI, the
fragments were methylated at G residues and bound to p53 (Fig. 9).
Methylation of G residues critical for p53 binding resulted in
interference with IP. For example, methylation at nucleotides, 217, 22,
227 to 229, and 233 of the 248 by insert from clone 11B3 completely
interfered with the binding of this fragment to p53 (Fig. 9, footprint 2).
When the opposite strand was analyzed, interference was observed at
the G residues corresponding to nucleotides 219, 223, 224, 230, 235, and
236 (Fig. 9, footprint 1). For DP, labelled DNA fragments were first
subject to IP, then incubated with various amounts of DNase I. For
clone N22, p53 binding provided protection against DNase I cleavage at




2070979
-37-
residues 187 to 211 (Fig. 9, footprint 9). MI showed interference by G
residues only within the region protected by DNase I (Fig. 9, footprint
10). Other examples of DP and MI mapping are shown in Figure 9.
p53-binding DNA fragments were subcloned and labeled on one end,
gel-purified and subjected to DP or MI mapping. For MI, 10 ng of DNA
were incubated in 200 ul of 50 mM Na-cacodylate, 1 mM EDTA, pH 8.0
and 5 ul of 1096 dimethylsulfate/9096 ethanol for 5 minutes at 20 ° C
to
methylate G residues. Fifty ul containing 1.5 m Na-acetate, 1 M
B-mercaptoethanol and 60 ug of glycogen were added. The mixture was
ethanol-precipitated, washed, and resuspended in 5 ul of 3mM Tris, 0.2
mM EDTA, pH 7.5, and allowed to bind to wild-type p53 as described in
the legend to Figure 1. After IP and DNA purification, the samples
were incubated with 100 ul of 1 M piperidine at 90 ° C for 30 minutes.
The samples were then dried under vacuum and separated
electrophoretically on a 696 polyacrylamide sequencing gel. The
control DNA samples were carried through all incubations except no
p53 was added. For these control samples, the protein A Sepharose
pellets were treated with SDS and proteinase K without removal of the
supernatants (which contained the labeled DNA in the absence of p53).
For DP assays, end-labeled DNA fragments were
immunoprecipitated as described in the legend to Figure 8. The protein
A Sepharose pellets were incubated for two minutes at 25 ° C with
200
ng DNase I in 5 mM MgCIZ. After purification of the DNA, as described
above, samples were separated by electrophoresis on sequencing gels
and loaded as described above for MI. MI was performed on all 18
genomic DNA fragments which bound to p53. DP assays were
performed on 13 fragments and the regions of protection uniformly
coincided with those indicated by the MI assays.
Example 10
This example analyzes the sequences of p53-bound DNA regions.
We next compared the sequences of the twenty-three clones.
The average insert sequence was 307 by (range 139-470). We found that
ten of the twenty-three clones were not unique, showing at least one
hundred contiguous nucleotides identical to one other clone. Thus, the
twenty-three clones represented only eighteen independent genomic




2070979
-38-
DNA fragments. We attempted to find similarities among these
eighteen fragments by computer methods, but found no significant
relationships. However, when we aligned the regions involved in p53
binding (as assessed by MI and DP), a striking and consistent feature of
the clones became apparent (Fig. 10). Each of the binding sites
contained two copies of the 10 by motif 5~-RRRCWWGYYY-3',
separated by 0 to 13 bp. One clone (5592) contained two separate areas
of footprinting, and both regions contained a dimer of the 10 by motif
(Fig. 10). In all clones, the regions displaying DP and MI were always
centered within the dimers, and G residues within the 10 by motif
strongly interfered with binding to p53 (examples in Fig. 9). The 10 by
consensus monomer contained an internal symmetry, with two
oppositely oriented half-sites of the form 5~-RRRCW-3~. This symmetry
was extended in the dimers, which contained four half-sites oriented in
alternating directions, forming a pseudopalindromie structure,
sometimes with an intervening loop. This consensus dimer was also
recognized in the p53 binding sequence mapped within plasmid CBE lOd
(Fig. 10). Despite the remarkable symmetry noted for all p53 binding
sequences, none of the genomic sites were palindromic.
Example 11
This example demonstrates that a dimer of the consensus motif
is required to bind p53 and that mutant p53 proteins found in tumors do
not bind to the consensus sequence.
To determine if the 10 by consensus monomer could bind to p53,
a synthetic oligonucleotide (5~-AGGCATGTCT-3~) containing the
consensus sequence was studied. Oligonucleotide duplexes were tested
either directly or after cloning into plasmid vectors. The monomer was
found not to bind to p53, either alone (not shown) or flanked by 43
nucleotides of plasmid sequences (Fig. i lA, lane 5). In contrast, the
dimers (composed of two copies of the monomer arranged in
head-to-head, tail-to-tail, or head-to-tail orientation), each bound
strongly to p53 protein (Fig. 11A, lanes 1 to 4, 6). Higher-order
oligomers of the monomer did not bind any better than the dimer in the
IP assay (Fig. 11A, lanes 6 to 8). A different monomer, still fitting the
consensus sequence, but perfectly palindromic, also bound as a dimer




2070979
-39-
but not as a monomer (Fig. 10, synthetic oligonucleotides 3,4). Two
variants of the consensus motif were also tested for binding. In the
first, the two critical G:C bp's at position 4 and 6 of the monomer were
substituted with A:T bp's (Fig. 10, synthetic oligonueleotides 1,2).
Although this sequence was perfectly symmetrical, it did not bind to
p53 either as a monomer or as a dimer. We also tested direct repeats
of the 5'-PuPuPuC(A/T)-3' half-site, and found that these did not bind
to p53 (Fig. 10, synthetic oligonucleotide 5). Thus, the mirror-image
symmetry of the half-sites within the 10 by consensus monomer was
critical for its activity.
Finally, we tested p53 mutants representing each of the four
"hot-spots" frequently altered in human cancers ((Nigro, et al. (1989),
Nature, 342:?05-T08, and Holstein, et al. (1991), Science, 253:49-53) for
ability to bind to the consensus dimer. None of the p53 mutants bound
appreciably to this sequence (Fig. 11B) under conditions where the wt
protein bound strongly. These experiments also showed that _in vitro
translated p53, as well as that purified from baculovirus-infected insect
cells, had the capacity to bind DNA specifically.
In summary, a set of human genomic DNA sequences which
could bind p53 were isolated and used to define a consensus binding
sequence for p53. The symmetry of the four half-sites within the
consensus dimers suggests that p53 interacts with DNA as a tetrameric
protein. This is consistent with studies suggesting that p53 assembles
into homotetramers (Krais, et al. (1988), J. Virol., 62:4?3?-4744, and
Weinberg (1991), Science, 254:1138-1146).
Example 12
This example demonstrates that intact p53 can activate
expression in human cells.
We first made reporter plasmids (PG"-CAT series) containing
part of the polyomavirus early promoter and the CAT gene located
downstream of DNA sequences which could bind to p53 in vitro (Figure
8). For the CAT reporters, concatemers of the p53-binding region of
CBE were formed by ligation of complementary oligonucleotides, ligated
into the EcoRV site of pBluescript II SK+ (Stratagene) to form the PG"
and MGn series. The BgIII-BamHI fragment of pPyOICAT (Murakami, et

CA 02070979 2003-07-03
-40-
al. (1990) Oncogene, 5:5), containing the polyomavirus early promoter and the
CAT
gene coding region, was ligated into the BamHI site of the PGn and MG" series
clones
to form the PG"-CAT and MG"-CAT series, and the orientation of the inserts
characterized by restriction enzyme analysis. The PG9-MG"-CAT and PG~3-MG"-
CAT series were formed by excising the HindIII-SaII fragments of PG9-CAT and
PG13-CAT, blunt-ending, attaching XbaI linkers, and ligating into the XbaI
site of the
MG"-CAT series plasmids (where n=1, 5, 10, and 15). For the yeast ~3-
galactosidase
reporter plasmids, PG and MG sequences were ligated as SaII-SmaI fragments to
the
Sail and filled-in XhoI sites of pCZ (Buchanan, et al. (1988), Mol. Cell
Biol.,
8:50806). The construction of the p53-wt expression construct has been
described
(Baker, et al. (1990), Science, 249:912); the mutant expression plasmids were
constructed similarly from the previously described cDNA plasmids (Nigro, et
al.
(1989), Nature, 342:705, and Kern, et al. (1991), Oncogene, 6:131), or in the
case of
the engineered phosphorylation site mutants, by in vitro mutagenesis (Altered
Sites,
Promega) with verification by sequencing. The construction of the yeast p53
expression vectors based on pRS314 has been described (Nigro, et al (1992)
Mol. Cell
Biol. 40 (12):1357-1365.
For the p53 binding sequences, we used a series of concatemers of the
oligonucleotide PG (5'-CCTGCCTGGACTTGCCTGG-3'). This contained the
binding region of plasmid CBE, previously shown to bind p53 in vitro. The
reporter
and an expression vector coding for the intact human wild-type protein (p53-
wt) (Fig.
12B), were transfected together into the human colorectal cancer cell line HCT
116.
This line makes low amounts of apparently wild-type p53 protein.
Exons 5-8 of the p53 genes from HCT 116 cells were amplified by PCR and
sequenced as described in Sidransky et at. (1991), Science, 252:706.
Previously,
these exons had been shown to contain over 90% of the mutations observed in
human
tumors. No mutations were observed from HCT 116 cells. Small amounts of
apparently wild-type protein could be detected in Western blots of HCT 116
protein
(See Fig. 16B).


CA 02070979 2003-07-03
-41 -
The intact wild-type p53 protein was indeed able to activate transcription
(Fig.
13). We found that the level of transactivation of the CAT gene depended on
the
strength of binding to p53 of the upstream sequences. Thus, the longer the
number of
PG repeats, the greater the binding to p53 in vitro (Fig. 13A) and the higher
the CAT
expression in vivo (Fig. 138, lanes 1-7). The level of transactivation also
increased
with the amount of p53 expression vector transfected (Table3).
,:
i
,,
,.


CA 02070979 2003-07-03
-41a-
Table 3. Transactivation in human cells
Relative CAT Activityl/
p53
Expression Exp. 2
Vectorz/ Reporter Exp.l .85wg 2.25~g Exp.3
p53-wt PG~3CAT 100 100 210 100
Tumor-derived mutations:
p53-143 PG,3CAT 3 1 4


p53-175 PG~3CAT 2 3 6


p53-248 PG13CAT 2 4 7


p53 -273PG13CAT 2 2 3


Endogenous activities:
p53-wt MG15CAT3/ <1 2
none4/ PG13CAT 20 2
1/ All transfections used 1.7 ~g reporter in HCT 116 cells. Transfection and
CAT assays were performed as described (Sadransky, et al. (1991), Science,
252:706). Activities reflect the fraction of chloramphenicol converted to an
acetylated form, expressed as relative values with one transfection of each
experiment
arbitrarily designated as having a value of 100. Three representative
experiments
(Exp.) shown.
Z/ Experiment 2 used both 0.85 and 2.55 pg levels of expressor. Experiment 1
used 1.7 pg, and experiment 3 used 0.85 pg.
3/ MG, 5-CAT provided a control for DNA-binding specificity and an estimate of
background from promoter-independent "readthrough" transcription and/or basal
promoter activity.
4/ The expressor-negative transfection provided an estimate of the activity
from
endogenous wild-type p53 of the HCT 116 cells. In various transfections, the
endogenous activity was 2-20% of that following transfection with exogenous
wild-
type p53, depending on the efficiency of transfection. The activity from
endogenous
p53 was diminished by transfection of oncogenic mutant p53.

CA 02070979 2003-07-03
-42-
Specificity for the binding sequences was tested by replacing six GC basepairs
in the
18-basepair oligonucleotide to generate a mutant form of PG, termed MG (5'-
CCTTAATGGACTTTAATGG-3'). This sequence, when multimerized and placed
upstream of the CAT reporter, did not bind to p53 in vitro (Fig. 14, lanes 7
and 8), nor
did it activate CAT expression in vivo (Table 3; Fig. 13B, lane 8; Fig. 15A,
lane 6,
Fig. 15B, lane 2). The transactivation of CAT was independent of the
orientation of
the PG multimer upstream of the CAT gene (Fig. 13B). Placing an additional 59
to
333 basepairs between the PG multimer and the promoter also had little effect
on
transactivation (Fig. 15A, lanes 2-5). However, placement of the PG binding
sequences downstream of the CAT gene did not allow transactivation (FIg. 15,
lane
3). These observations indicated that the PG sequences act as an upstream
activator
element, although not as a classical enhancer.
CAT assay. Cultures of HCT 116 cells at 50-80% confluence in 25 cmz flasks
were transfected using LipofectinT"" (BRL, Gaithersburg, MD) according to the
manufacturer's instructions. All flasks within an experiment were transfected
with
the same total amount of plasmid using pCMVneoBam or pBluescript II SK + as
"filler". Cells were harvested at 20-24 hr. and the CAT activity of the
lysates
measured by acetylation of 14C-labeled chloramphenicol (ICN) as previously
described (Gorman, et al. (1982), Mol. Cell Biol., 2:1044). The Bio-Rad
protein
assay was used to assure equivalence of lysate protein. Percent conversion to
the
acetvlated form of chloramphenicol was calculated



2070979
-43-
of ter quantitation by scintillation counting of excised chromatographic
spots. Results reported are representative of at least two transfections
done on separate days.
Example 13
This example demonstrates that oncogenic mutant p53 genes
consistently fail to transactivate.
The precise correlation between the binding of the PG multimer
in vitro (Fig. 13A) and the expression from PGn-CAT reporters in vivo
(Fig. 13B) strongly suggested that this expression was a direct result of
p53 binding to PG multimers in vivo. If such an activity of p53 were
crucial to its tumor suppressor activity, one would expect that
naturally occurring mutants of p53 would be detective in this function.
p53 mutations generally occur in four different p53 "hot-spot" regions,
representing four evolutionarily conserved domains of the p53 protein
(Baker, et al. (1989), Science, 244:21?, Hollstein, et al. (1991), Science,
. 253:49, and Soussi, et al. (198?), Onco~tene, i:?1). Mutant p53 genes
representing each of these hot-spots were transiently transfected into
cells together with the PG13-CAT reporter plasmid. The consistency
of the results obtained with these mutant expression vectors was
striking. All tumor-derived mutations examined had lost the ability to
transaetivate CAT, despite the wide range of positions (codons 143-2?3)
in which these mutations were situated (Table 3, and Figure 16A). As
might be expected for a cell line expressing low amounts of endogenous
wild-type p53, there was of ten some innate transactivating ability
associated with the HCT 116 cells themselves (Table 3; Fig. 13B, lane i;
Figure 16A, lane 1); interestingly, the levels of CAT observed with the
mutants (Table 3 and Fig. 16A, lanes 4-?) were actually less than that
observed with no exogenous p53 expression vector, suggesting that the
mutant p53 products might be inhibiting the low amounts of expression
mediated by the endogenous p53 (discussed below). The mutants studied
included one which was typical of those found in tt~e germline of
Li-Fraumeni patients (248trp)(Malkin, et al. (1990), Science. 250:1233;
Strivastava, et al. (1990), Nature. 348:?4?) and three found commonly in
a variety of human tumors (143°al, i?5hi$, 2?3hi°)(Hollstein, et
al.
(1991), Science, 253:49). Immunoblots confirmed that mutant p53




20709?'9
-44-
proteins were expressed at levels comparable to those of wild-type p53
(Fig. 16B).
Western blots were performed as follows: 100 ug protein of
each cell lysate was separated on a .1096 polyacrylamide SDS gel,
transferred onto a PVDF (Millipore, Bedford, MA) membrane, blocked
with 596 nonfat milk, incubated with 1 ug/ml PAb1801 (Oncogene
Sciences) then i2sl_labeled goat-anti-mouse Ab (NEN), and
autoradiographed. The inability of p53 proteins having tumor-derived
mutations to bind specifically to the p53 recognition sequences in this
construct (Fig. 14, lanes 3-6) is thus consistently reflected as defects in
transactivation.
Example 14
This example demonstrates that intact p53 transactivates in
yeast.
We next sought to determine whether this activity of p53 was
confined to mammalian cells. If the ability of p53 to bind to DNA
sequences in vivo and activate the transcription of adjacent genes were
an intrinsic feature of the protein, it was possible that this activity
would be manifest in simpler eukaryotes. Indeed, the N-terminal acidic
activation domain of p53 has been reported to function in _S. cerevisiae
when fused to the DNA-binding domain of GAL4 (Fields, et al. (1990),
Science, 249:1046). We thus stably transfected yeast with the LacZ
reporter gene placed downstream of PG multimers (PGn-LacZ), as well
as with a galactose-inducible p53 expression vector (Fig. 12). The
addition of galactose to the medium resulted in p53 expression
accompanied by a dramatic elevation in S-galactosidase expression
(Table 4). When the MG multimer (non-p53 binding) was substituted for
the PG multimer, no activation was observed (Table 4). Moreover, p53
mutants had no capacity to transactivate in yeast cells (Table 4). Thus,
the results in S. cerevisiae were analogous to those observed in human
cells.




-45- 2070979
Table Activation of Gene Expressionin Yeast p53
!. by


p53 B-Gal Activity


Expression


Vector Reporter Exp 1 Exp 2



none PG16-LacZ 3


Yp53 PG16-LacZ 13,000 8,000


Yp53-143 PG16-LacZ 150 85


Yp53-273 PG16-LacZ 5 2


Yp53 PG4-LacZ 15,000 11,000


Yp53 PGl-LacZ 2,600 3,000


Yp53 MG15-LacZ 15 4


Wild-type or mutant p53 expression vectors and the
B-galactosidase reporter plasmids were transfected into _S.
cerevisiae and clones obtained. p53 expression was induced
with galactose, and 8-galactosidase activity was measured in
units of nanomoles per minute per milligram of protein. Two
independent clones (Exp. 1 and Exp. 2) were tested. Less than
1 unit was seen in the absence of galactose induction. The
residual activity of the Yp53-143 mutant may have been due to a
slight wild-type activity observable with valine to alanine
substitution mutants at the relatively low temperature (30°C)
used for yeast growth.
Example 15
This example demonstrates the dominant-negative effect of p53
mutations on wild-type p53's ability to bind DNA and enhance
transcription.
If the transcriptional activation of p53 were fundamental to its
biological role, p53 mutants should be able to interfere with this
activation, just as p53 mutants can interfere with the tumor-suppressor
activity of wild-type p53 when both are expressed (Levine, et al.
(1991), Nature, 351:453). To assess this possibility, we cotransfected
the wild-type and mutant p53 genes with the PG13-CAT reporter
plasmid into HCT 116 cells (Figs. iZ and 18). When equal amounts of
wild-type and mutant p53 expression vectors were used, the expression




2070979
-46-
of CAT. decreased by approximately 5096 compared to that achieved by
wild-type p53 alone. A three-fold ratio of mutant p53 to wild-type p53
produced an 84-9596 reduction. When additional wild-type p53 was
substituted for mutant p53 in the cotransfection experiment, the CAT
expression increased rather than decreased, as expected (Fig. 18, ~~wt~~
lane). Thus, expression of the CAT reporter was dependent on the
ratio of wild-type to mutant p53 in the cell.
The dominant-negative effect could be caused by a failure of the
mutant/wild-type complexes to bind to DNA or to a failure to activate
transcription once bound. To distinguish between these two
possibilities, we co-translated wild-type p53 and the lTShis mutant of
p53 in an experiment similar to that shown in Figure 14. The mutant
p53 protein inhibited the ability of wild-type protein to bind DNA by
over 9096 when co-expressed at a 3:1 ratio.



Image


Image

Representative Drawing

Sorry, the representative drawing for patent document number 2070979 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date 2006-01-03
(22) Filed 1992-06-10
(41) Open to Public Inspection 1992-12-15
Examination Requested 1999-05-28
(45) Issued 2006-01-03
Expired 2012-06-10

Abandonment History

Abandonment Date Reason Reinstatement Date
2005-06-10 FAILURE TO PAY APPLICATION MAINTENANCE FEE 2005-08-30

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $0.00 1992-06-10
Registration of a document - section 124 $0.00 1993-07-30
Registration of a document - section 124 $0.00 1993-07-30
Registration of a document - section 124 $0.00 1993-07-30
Registration of a document - section 124 $0.00 1993-07-30
Maintenance Fee - Application - New Act 2 1994-06-10 $100.00 1994-03-24
Maintenance Fee - Application - New Act 3 1995-06-12 $100.00 1995-03-23
Maintenance Fee - Application - New Act 4 1996-06-10 $100.00 1996-03-22
Maintenance Fee - Application - New Act 5 1997-06-10 $150.00 1997-03-26
Maintenance Fee - Application - New Act 6 1998-06-10 $150.00 1998-03-19
Maintenance Fee - Application - New Act 7 1999-06-10 $150.00 1999-03-22
Request for Examination $400.00 1999-05-28
Maintenance Fee - Application - New Act 8 2000-06-12 $150.00 2000-04-13
Maintenance Fee - Application - New Act 9 2001-06-11 $150.00 2001-04-10
Maintenance Fee - Application - New Act 10 2002-06-10 $200.00 2002-05-10
Maintenance Fee - Application - New Act 11 2003-06-10 $200.00 2003-05-26
Back Payment of Fees $200.00 2003-08-25
Maintenance Fee - Application - New Act 12 2004-06-10 $250.00 2004-06-07
Final Fee $300.00 2005-07-29
Reinstatement: Failure to Pay Application Maintenance Fees $200.00 2005-08-30
Maintenance Fee - Application - New Act 13 2005-06-10 $250.00 2005-08-30
Maintenance Fee - Patent - New Act 14 2006-06-12 $250.00 2006-05-05
Maintenance Fee - Patent - New Act 15 2007-06-11 $450.00 2007-05-07
Maintenance Fee - Patent - New Act 16 2008-06-10 $450.00 2008-05-12
Maintenance Fee - Patent - New Act 17 2009-06-10 $450.00 2009-05-19
Maintenance Fee - Patent - New Act 18 2010-06-10 $450.00 2010-05-17
Maintenance Fee - Patent - New Act 19 2011-06-10 $450.00 2011-06-10
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
THE JOHNS HOPKINS UNIVERSITY
PHARMAGENICS, INC.
Past Owners on Record
KINZLER, KENNETH W.
SHERMAN, MICHAEL I.
VOGELSTEIN, BERT
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 1999-07-07 54 2,521
Description 2003-07-03 53 2,512
Claims 2003-07-03 6 249
Description 1994-04-23 48 2,579
Cover Page 1994-04-23 1 21
Abstract 1994-04-23 1 13
Claims 1994-04-23 7 305
Drawings 1994-04-23 24 646
Description 2004-12-17 53 2,516
Claims 2004-12-17 6 249
Cover Page 2005-12-20 1 27
Assignment 1992-06-10 23 878
Prosecution-Amendment 1999-05-28 4 155
Prosecution-Amendment 2003-01-03 3 110
Prosecution-Amendment 2003-07-03 24 1,064
Prosecution-Amendment 2003-08-25 2 46
Prosecution-Amendment 2004-07-05 2 45
Fees 2004-06-07 1 49
Prosecution-Amendment 2004-12-17 5 193
Correspondence 2005-07-29 1 50
Fees 2005-08-30 2 114
Fees 2005-08-25 2 81
Fees 1997-03-26 1 67
Fees 1996-03-22 1 50
Fees 1995-03-23 1 65
Fees 1994-03-24 1 43